EP0494955B1 - Cell-free synthesis and isolation of novel genes and polypeptides - Google Patents

Cell-free synthesis and isolation of novel genes and polypeptides Download PDF

Info

Publication number
EP0494955B1
EP0494955B1 EP90915566A EP90915566A EP0494955B1 EP 0494955 B1 EP0494955 B1 EP 0494955B1 EP 90915566 A EP90915566 A EP 90915566A EP 90915566 A EP90915566 A EP 90915566A EP 0494955 B1 EP0494955 B1 EP 0494955B1
Authority
EP
European Patent Office
Prior art keywords
polysomes
sequence
mrna
interest
rna
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
EP90915566A
Other languages
German (de)
French (fr)
Other versions
EP0494955A4 (en
EP0494955A1 (en
Inventor
Glenn Kawasaki
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Optein Inc
Original Assignee
Optein Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Optein Inc filed Critical Optein Inc
Publication of EP0494955A1 publication Critical patent/EP0494955A1/en
Publication of EP0494955A4 publication Critical patent/EP0494955A4/en
Application granted granted Critical
Publication of EP0494955B1 publication Critical patent/EP0494955B1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/02Acyclic radicals, not substituted by cyclic structures
    • C07H15/12Acyclic radicals, not substituted by cyclic structures attached to a nitrogen atom of the saccharide radical
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1041Ribosome/Polysome display, e.g. SPERT, ARM
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/6867Replicase-based amplification, e.g. using Q-beta replicase

Definitions

  • the present invention generally relates to the synthesis and isolation of novel genes and polypeptides in vitro and, more specifically, to methods of generating and expressing semi-random DNA or RNA sequences, isolating novel genes from those sequences, and using those genes to create novel polypeptides.
  • novel genes and polypeptides from semi-random sequences is currently limited by the need to screen a large, genetically diverse population of cells in order to obtain the sequence(s) of interest.
  • a polypeptide string of 10 amino acids has 20 10 or approximately 10 13 possible permutations. If 10 of these permutations had a desirable characteristic (such as the ability to bind a specific antigen), then a population of 10 12 would have to be screened for the expectation of finding one desirable novel gene.
  • the screening of a large number of new sequences for a specific property is virtually unfeasible, unless the novel gene provides the organism with a distinct growth or survival advantage. Indeed, under the current state of the art, the 10 12 independently transformed microorganisms would have to be screened individually to locate that one desirable novel gene.
  • colonies derived from each transformed cell must be treated to break open the cells.
  • 1000-2000 bacterial colonies per standard petri dish are lysed (e.g., by chloroform) for the screening procedure.
  • 500,000 to 1 billion petri dishes would be necessary.
  • 10,000 to 100,000 liters of logarithmically dividing cells may be necessary for producing the large numbers of transformable cells.
  • a gene product may be secreted and attached to the outside of a cell, it may be detected by its ability to bind a fluorescent compound or other marker.
  • cell sorters may be used to screen for the synthesis of a novel desirable polypeptide.
  • flow rate 5,000 cells per second, it would take a cell sorter over 60 years to screen 10 12 cells.
  • present day screening methods which are both extremely costly and time-consuming, effectively prohibit the isolation of novel genes and polypeptide from semi-random sequences.
  • the present invention describes a method for cell-free screening of novel genes and polypeptides. This method avoids the problems associated with large numbers of transformed organisms as well as the limitations of the method disclosed by Fields and Song, and may be completed within a few weeks. Therefore, the methodology allows a substantial time and monetary saving in the isolation of novel gene products.
  • the present invention relates to methods for synthesizing, screening, and selecting high numbers of novel genes and polypeptides.
  • a method for isolating a nucleotide sequence which encodes a polypeptide of interest comprising transcribing or replicating an in vitro expression unit which comprises a 5' untranslated region containing a RNA polymerase binding sequence, a ribosome binding sequence, a translation initiation signal, and one or more semi-random nucleotide sequences to produce a mRNA library; translating the mRNA library under conditions which maintain polysomes having polypeptide chains attached thereto; contacting the polysomes to a substance of interest that is capable of specifically reacting with the polypeptide of interest and isolating mRNA from polysomes that specifically react with the substance of interest.
  • the process may be repeated on mRNA that has been enriched for desirable sequences by amplifying the RNA or respective cDNA. Subsequently, this amplified subset of genes may be cycled through the various steps outlined above to further enrich for desirable novel genes until desirable sequences represent a significant (>10 -3 ) fraction of the truncated population. In principle, the method may be repeated until the population of genes is nearly homogeneous.
  • a method for producing novel polypeptides by expressing the nucleotide sequence isolated in the method of the first aspect comprising, subsequent to the contacting step of the first aspect, isolating said polysomes that bind to said substance of interest; disrupting said isolated polysomes to release mRNA; recovering said mRNA; constructing cDNA from said recovered mRNA; and expressing said cDNA to produce novel polypeptides.
  • a method for producing novel polypeptides by expressing the nucleotide sequence isolated in the method of the first aspect wherein, during said translating step, the conditions optionally permit polypeptide chains to be released from the polysomes, and further comprising isolating the mRNA encoding the polypeptides identified in the contacting step by subdividing, repeating the steps of transcribing, translating, contacting and isolating by subdividing, such that the gene of interest is isolated; constructing cDNA from said isolated gene; and expressing said cDNA to produce novel polypeptides.
  • the expression unit described above comprises an RNA polymerase binding sequence, a ribosome binding site, and a translation initiation signal.
  • the expression unit may further comprise a translation enhancer or "activator" sequences, a 3' tail of a selected sequence and appropriate restriction sites.
  • the semi-random DNA sequences may be generated by mechanically, chemically, or enzymatically fragmenting naturally-occurring DNA, by chemically synthesizing the DNA, or by polymerizing the DNA directly onto the expression unit.
  • the substance of interest may be a surface antigen, receptor protein, toxin, organic polymer, active site of a protein molecule, metabolite, antibody, metal, hormone, or other compound.
  • novel genes may have virtually infinite diversity and may code for new polypeptides with commercially important properties, such as novel catalytic activities or the ability to bind selectively to specific substances.
  • Novel genes may be constructed which comprise open reading frames from existing genes or from semi-random nucleotide sequences of chemically synthesized DNA. They may be expressed in a wide variety of organisms using existing promoters, enhancers, initiation codons, plasmids, ribosomal binding sites, and/or terminators. In some cases, it may be advantageous to express the novel genes in vitro, as part of a large-scale production process.
  • the present invention describes a multistep process for constructing and isolating novel genes and gene fragments which encode novel polypeptides with specific binding and/or biological activities.
  • the process comprises the following steps:
  • steps 3-8 repetition of steps 3-8 is preferable to further increase the frequency of specific binding proteins above a background of nonspecific binding of polysomes.
  • the isolated, purified novel gene(s) produced by the methods described herein are capable of generating a variety of polypeptide(s) of interest using standard expression techniques, as positive proof that the gene codes for the desired product.
  • DNA and/or polypeptide sequencing by conventional methods may be used to identify the composition of the novel polypeptide.
  • large-scale production of the novel polypeptide(s) may be accomplished by chemical synthesis (if the amino acid sequence is relatively short) or through recombinant DNA methods, using genetically engineered microorganisms. Alternatively, large-scale in vitro transcription and/or translation methods may be used to produce commercial quantities of the polypeptide.
  • the DNA sequence coding for the selected polypeptide may also be incorporated into larger genes (i.e., such as into the hypervariable regions of antibody genes) to create hybrid proteins with the specific binding and/or biological activities of the originally isolated novel polypeptides, in addition to other binding and biological activities.
  • the expression unit comprises a 5' untranslated region and may additionally comprise a 3' region.
  • the 5' untranslated region of the expression unit contains a promoter or RNA polymerase binding sequence, a ribosome binding sequence, and a translation initiation signal.
  • the 5' untranslated region (“head") may also contain convenient restriction sites and a translation enhancer or "activator” sequence(s).
  • the 3' region may contain convenient restriction sites and a 3' tail of a selected sequence.
  • the expression unit may be chemically synthesized by protocols well known to those skilled in the art. Alternatively, these elements may be incorporated into one or more plasmids, amplified in microorganisms, purified by standard procedures, and cut into appropriate fragments with restriction enzymes before assembly into the expression unit.
  • the 5' untranslated region contains a promoter or RNA polymerase binding sequence.
  • High-efficiency promoters such as those for the T7, T3, or SP6 RNA polymerase, are preferred in this invention for the following reasons.
  • Such promoters are short DNA sequences of known composition, are extremely specific for their relative polymerases, and are highly active, allowing for greater than 50 rounds of transcription per DNA template.
  • T7, T3, and SP6 polymerases are commercially available from many sources and are components of well-characterized transcription kits.
  • the consensus sequence is TAATACGACTCACTATAGGGAGA (23 base pairs).
  • a DNA sequence which codes for a ribosomal binding site Positioned downstream of or within the promoter region is a DNA sequence which codes for a ribosomal binding site.
  • This ribosome binding site may be specific for prokaryotic ribosomal complexes (including ribosomal RNAs) if a prokaryotic translation procedure is used.
  • a preferred embodiment of this invention uses a eukaryotic sequence and an in vitro eukaryotic translation system, such as the rabbit reticulocyte system (Krawetz et al., Can. J. Biochem. Cell. Biol. 61: 274-286, 1983; Merrick, Meth. Enzymol. 101 :38, 1983).
  • a consensus translation initiation sequence, GCCGCCACCATGG, as well as other functionally related sequences have been established for vertebrate mRNAs (Kozak, Nucleic Acids Res. 15 :8125-8148, 1987). This sequence or related sequences may be used in the novel gene construction to direct protein synthesis in vitro.
  • the ATG triplet in this initiation sequence is the translation initiation codon for methionine; in vitro protein synthesis is expected to begin at this point.
  • Appropriate restriction sites may also be included in the expression unit to assist in future genetic engineering.
  • the sextuplet, CCATGG is the recognition sequence for the restriction endonuclease, NcoI.
  • a NcoI "cutting site" positioned downstream of the ribosomal binding site is a convenient splice point for subsequent genetic engineering.
  • the expression unit may be spliced from the novel gene at this site, and another promoter may be attached for expression in vivo and large-scale production of the novel polypeptide.
  • the NcoI site may also be used as a convenient cloning site for the construction of hybrid proteins, where two different polypeptide domains are brought together and expressed as a single protein.
  • a DNA sequence with at least one restriction endonuclease site for subsequently cloning the novel gene into plasmids.
  • the octameric sequence, GCGGCCGGC is recognized by NotI nuclease and is particularly useful because it would rarely fall within the novel coding region of the gene (NotI is expected to cut totally random DNA once every 65,536 base pairs).
  • Other restriction sites may also be used; the expected frequency of cutting the novel coding region is dependent upon the nucleotide composition or the DNA source of the coding region. It should be noted that certain palindromic sequences may interfere with translation; however, some sequences may also enhance the rate of translation.
  • the expression unit may also comprise a 3' region. It is desirable to construct known 3' regions (tails) with palindromic sequences for at least two reasons. First, 3' restriction sites would be convenient for any later genetic engineering of the polypeptide coding region. For example, if NotI sites were located in both the 5' and 3' regions, a desirable polypeptide coding sequence could be cut out with NotI "sticky ends" for further cloning. Second, palindromes may cause secondary structures which impede translocation, thus, palindromes in the 3' region may slow down the movement of ribosomes during translation.
  • the 3' region may also contain a poly-A or other polynucleotide stretch for later purification of the mRNA from other components in the in vitro translation reaction by hybridization to a complementary homopolymeric sequence.
  • the expressed polypeptides contain both nonrandom and semi-random amino acid sequences.
  • the nonrandom component of the coding region is synthesized and produced with the nonrandom 5' untranslated region and/or with the 3' region.
  • This nonrandom coding sequence specifies a string of amino acids (an identification or "ID" peptide) that is conserved among the billions of novel polypeptides.
  • ID peptide would be useful for quantifying the amount of novel polypeptide and for purification of the novel polypeptide (given that an antibody against the ID peptide is available or can be produced).
  • Substance P 11 amino acid Substance P
  • Anti-Substance P antibodies are commercially available for detecting and quantifying fusion proteins containing Substance P.
  • Another example is the eight amino acid marker peptide, "Flag" (Hopp et al., Bio/Technology 6 :1204-1210, 1988).
  • Amino-terminal ID peptides have at least two advantages over carboxy-terminal ID peptides. First, it is easier to make gene constructions which maintain the proper reading frame of the N-terminal ID, because long stretches of semi-random DNA or RNA will tend to end in all three reading frames for a C-terminal ID. Second, the N-terminal ID may be designed to function as a signal peptide in a transformed organism, allowing for the possible secretion of the novel polypeptide during large-scale production.
  • C-terminal ID polypeptides may also be used.
  • One preferable C-terminal polypeptide is polyglycine, which is encoded by poly-dG and is read Gly-Gly-Gly, etc., regardless of the reading frame of the semi-random sequences.
  • the polyglycine 3' end of the polypeptide may act as a noninterfering tether of the nascent peptide and allow the semi-random sequences greater access to bind molecules of interest.
  • the poly-dG sequence may be used for priming second strand synthesis of the cDNA and may be useful for purification of the RNA or DNA with polyC or poly-dC.
  • GGGCGGGC... may be used to code for a recognizable peptide sequence which is expressed in all reading frames.
  • a preferable form of the ID peptide is one which may be cleaved from the novel polypeptide by simple chemical or enzymatic means.
  • an RNA expression unit may be constructed for semi-random polypeptide synthesis.
  • One possible advantage of the RNA expression unit is that the recovery of the polysomal mRNA does not have to go through an initial cDNA stage. Instead, the mRNA with the desired sequences may be amplified with an RNA-directed RNA polymerase, such as that of QB (Q Beta) replicase (Haruna and Spiegelman, Proc. Nat. Acad. Sci. 54: 579-587, 1965). This enzyme can make one billion copies of recombinant RNA in 30 minutes (Lizardi et al., Bio/Technology 6 :1197-1202, 1988).
  • QB Q Beta replicase
  • RNA libraries capable of in vitro replication may be created which contains the semi-random gene sequences.
  • RNA expression unit may be constructed by chemically synthesizing the appropriate RNA molecules and assembling them via an RNA ligase, such as the T4 RNA ligase (commercially available), which links together single-strand RNA and/or single-strand DNA.
  • an RNA ligase such as the T4 RNA ligase (commercially available)
  • RNA expression units and semi-random sequences are attached to the expression unit. Since the RNA expression units and semi-random sequences may be generated from a DNA template or constructed from chemically synthesized RNA or mRNA fragments in much the same manner as DNA expression units, the following description merely describes the process for semi-random DNA attachment to the expression unit. Those skilled in the art will readily be able to construct the RNA-equivalent of the expression units attached to semi-random polynucleotides.
  • Semi-random DNA may be generated by at least three methods.
  • First, naturally-occurring DNAs from virtually any living source may be mechanically, chemically, or enzymatically fragmented and attached to the 5' untranslated region with DNA ligase. Mixtures of fragments from different DNA sources may be used.
  • the end result may be the selectable expression of an active "open reading frame"--a portion (fragment) of a protein that has no "nonsense" (or "stop") codon, unless the activity resides in the extreme C-terminus of the molecule.
  • a gene coding for a known function may be fragmented; the resulting pieces are ligated to the 5' untranslated region and later screened for the expression of activity in the polysome assay.
  • Gene fragment analysis may be useful for creating small biologically active peptides and hybrid therapeutic proteins and may be beneficial for drug delivery, if smaller size assists the peptide in reaching the target site.
  • the "fragmented" DNAs may be semi-randomly sized cDNA molecules from a cDNA library.
  • cDNAs in vitro and using polysome selection, a very rare partial or perhaps even full-sized gene may be isolated through binding the polysome to antibody, receptor protein, or other diagnostic molecule.
  • the cell-free expression of cDNA "fragments" as herein described may be orders of magnitude more sensitive than previously described methods in locating desirable cDNA clones.
  • a second method for generating semi-random DNA is to chemically synthesize the DNA.
  • relatively long DNA molecules of approximately 100 nucleotides may be synthesized with mixtures of nucleotides at each position.
  • a statistical problem of nonsense codons becomes apparent with chemically synthesized DNA.
  • an active, open reading frame is located from within existing protein sequences. "Open reading frame” implies that no stop codon exists and often indicates a sequence from within a protein coding region.
  • the probability of at least one stop codon within that string is about 76%. Stop codons cause termination of translation and release of the nascent polypeptide from the ribosome complex. Therefore, strategies to reduce the frequencies of nonsense codons and to bypass the usual result of nonsense codons during protein translation are preferable, and discussed below.
  • the A, T, C, and G base composition may be manipulated to favor certain codons and in particular to reduce the likelihood of nonsense codons.
  • the third position of each triplet codon may be synthesized with only C and T to theoretically avoid nonsense codons.
  • Lim and Sauer Nature 339 :31-36, 1989
  • Lim and Sauer have used an equal mixture of all four bases in the first two codon positions and an equal mixture of C and G at the third codon position in synthesizing new regions of lambda repressor.
  • the likelihood of at least one TAG stop codon is about 61%.
  • unequal mixtures of the bases are used in all three codon positions to reduce the frequency of stop codons, while still allowing a high frequency of all 20 amino acids at all codons.
  • first codon position equal molar amounts of C, A, and G are used, but only half that amount of T is used.
  • the amount of A is reduced to half of the level of the other three bases.
  • the third codon position only G and C or G and T are used, and in equal molar amounts.
  • the result of this strategy is a greater than 79% probability that no stop codons will be present in a string of 30 amino acids.
  • the proportions of the individual amino acids are slightly distorted in this case relative to a totally random DNA strategy. However, only tyrosine will be represented at less than half of the expected frequencies compared to the random situation.
  • nonsense suppressing tRNAs be used in the in vitro translation steps.
  • a nonsense suppressor which recognizes TAG (actually UAG in the mRNA) and inserts tyrosine into the growing polypeptide chain is most desirable.
  • Such tyrosine-inserting nonsense suppressors may be generated by changing the anticodon region of a tyrosyl-tRNA in such a manner that the tyrosyl-tRNA now "reads" UAG instead of the normal UAU and UAC tyrosine codons in mRNA.
  • Normal tyrosyl-tRNAs will also be included in the translation step to read the tyrosine codons.
  • Nonsense suppressors can also be made for the other two nonsense codons.
  • tryptophane- or leucine-inserting suppressors of the UGA stop codon have been well characterized--as have many other nonsense suppressors.
  • the nucleotide sequences of many nonsense suppressors are known; and, therefore, the construction of such molecules would be evident to those skilled in the art.
  • Nonsense suppressors of mammalian translation systems are known (Burke and Mogg, Nucleic Acids Res. 13: 1317-1326, 1985; Capone et al., EMBO J. 4 :213-221, 1985; Diamond et al., Cell 25: 497-506, 1981; Hudziak et al., Cell 31: 137-146, 1982; Laski et al., EMBO J. 3 :2445-2452, 1984).
  • Geller and Rich have successfully suppressed nonsense codons in reticulocyte systems with yeast suppressor tRNAs and with bacterial suppressor tRNAs and tRNA synthetase. Therefore, the use of tRNA suppressors in the present invention to reduce premature release of polypeptides from the ribosomes during the translation step is well within the state of the art. Furthermore, both Pelham (ibid., 1978) and Geller and Rich (ibid., 1980) describe high levels of naturally-occurring nonsense suppression in eukaryotic translation systems.
  • Pelham shows that a particular UAG codon in tobacco mosaic virus may be "read” (suppressed) nearly 40% of the time by "supraoptimal concentrations of Mg +2 ", or a reported 2.1 mM MgCl 2 .
  • This level of magnesium ion or higher may therefore be used advantageously within the present invention to increase the readthrough of nonsense codons and to thereby reduce the problem of translation termination of longer semi-random nucleotide sequences.
  • different mixtures of bases at selected codon positions may be used to strongly bias in favor of or against a particular amino acid. For example, the elimination of G at position three in a codon prevents methionine and tryptophan from being included in the peptide.
  • a nucleotide mixture which is biased toward a high-cysteine content may be desirable for producing short peptides with internal disulfide bonds for structural rigidity. Such rigid peptides may bind other molecules more tightly.
  • Second-strand synthesis of these artificial nucleotide sequences may be accomplished by "random priming" and extension with DNA polymerase and/or by including a poly-dX tail from which to prime with poly-dX'. Other methods, such as the use of terminal palindromes that create "hairpin loops" for self-priming, may be used for second strand synthesis.
  • 100 ⁇ g of double-stranded DNA of 100 nucleotides contains about 10 15 molecules. If the semi-random synthesis strategy is used, the expectation is that each of these molecules codes for a different polypeptide. Therefore, a very large diversity in coding potential exists within laboratory bench-scale amounts of DNA.
  • Such a synthetic DNA molecule of 100 nucleotides is merely provided for purposes of illustration; longer sequences may also be synthesized.
  • shorter synthetic molecules may be generated and ligated together to make semi-random sequences of any given length. Shorter molecules are expected to preserve the reading frame of the synthetic DNA better than longer molecules, because each addition of chemically synthesized base is not 100%. Therefore, more nonsense codons may be avoided by the use of shorter artificial DNA molecules.
  • T4 RNA ligase or other means may be used to link together the short single-stranded DNAs.
  • a third method for generating semi-random DNA is to polymerize the molecules directly onto the 3' end of the 5' untranslated region. If no N-terminal ID sequence is used, the polymerization may occur immediately after the ATG initiation sequence or preferentially after the ATGG sequence--which preserves both the consensus vertebrate initiation site and the NcoI site.
  • the most commonly used enzyme for this polymerization is terminal transferase (usually from calf thymus), which is routinely used for generating homopolymeric regions for DNA cloning.
  • terminal transferase usually from calf thymus
  • semi-random heteropolymers of DNA may be synthesized on a DNA primer with a free 3'-OH.
  • the A, T, C, and G base composition may be manipulated to favor certain codons and reduce the frequencies of nonsense codon by controlling the relative concentrations of the four deoxynucleotide triphosphates.
  • a lower amount of dATP should reduce the frequencies of nonsense codons (TAA, TAG, and TGA).
  • E. coli DNA polymerase I is reported to carry out non-template (de novo) synthesis of DNA and may be used instead of terminal transferase (A. Kornberg, DNA Replication, W.H. Freeman & Co., San Francisco, Calif., 1980).
  • Other enzymes or chemical methods may also polymerize DNA directly onto the expression units. Second-strand synthesis is most easily accomplished by random primer extension, but other methods may provide the same result.
  • the use of nonsense suppressing tRNAs may greatly assist in overcoming the problem of stop codons in this semi-random DNA sequence.
  • RNA polymerase RNA polymerase
  • T7, T3, and SP6 RNA polymerases are commercially available and extremely active.
  • a DNA expression unit with a T7 promoter is treated with T7 RNA polymerase according to manufacturers' specifications. Approximately 50 mRNA copies may be synthesized routinely for each DNA molecule in 30 minutes. The DNA may be degraded with RNase-free DNase. If the original DNA library had a sequence diversity of 10 12 molecules, the resulting mRNA pool should reflect the same level of diversity but now contain 50 or more RNA copies of each different DNA molecule.
  • RNA library of 6 ⁇ g may contain 50 copies of 10 12 different mRNAs that are each capable of expressing a semi-random polypeptide of 30 amino acids. Since 6 ⁇ g is easily manageable in small test tubes, standard laboratory tools and vessels may be used.
  • the 5' ends of mRNAs need to be modified with the addition of diguanosine triphosphate "caps" (or analogs) for efficient translation in eukaryotic systems.
  • the 5' capped mRNA may be generated during in vitro transcription (Hope and Struhl, Cell 43: 177-188, 1985) and/or in the in vitro translation process (Krieg and Melton, Nucleic Acids Res. 12:7057-7070, 1984).
  • an excess of diguanosine triphosphate or an analog thereof is used during the RNA polymerization relative to GTP.
  • An mRNA capping kit based on this method is commercially available from Stratagene (California), which claims that 90%-95% of the resulting RNA is capped.
  • RNA-based such as the QB replicase system
  • T7 or other promoter systems see Lizardi et al., ibid, 1988
  • RNA-directed RNA polymerase is capable of making a virtually unlimited number of copies of the RNA library (one billion copies are easily attainable).
  • the diversity of the library remains the same.
  • RNA phages such as QB
  • the library may be self-sustaining at the RNA level without the necessity of going through a DNA intermediate.
  • in vitro translation methods are widely known. For convenience, the rabbit reticulocyte or wheat germ systems may be used with minor modifications.
  • In vitro translation kits are available commercially.
  • the "Translation Kit, Reticulocyte, Type I" from Boehringer Mannheim Biochemicals has all components for 100 translation reactions. Each reaction has been optimized for approximately 1 ⁇ g of mRNA in a 25 ⁇ l volume.
  • One ⁇ g of mRNA is sufficient to code for over 4 x 10 12 novel genes, as described above. Therefore, it is possible to translate extremely high numbers of novel genes in relatively small volumes. For example, 10 13 80S ribosomes only weigh approximately 66 ⁇ g. Because of the small size of the mRNA, only a few ribosomes per message are expected to saturate the mRNAs.
  • GTP and m7G(5')ppp(5')G are required for the efficient translation of in vitro transcribed RNA.
  • diguanosine triphosphate (or analog thereof) and guanylyltransferase may be necessary for capping the RNA molecules during translation.
  • ribonuclease inhibitors such as heparin.
  • Eukaryotic systems such as the wheat germ and reticulocyte translation methods, may yield similar results to prokaryotic systems.
  • the prokaryotic systems have the advantages of smaller ribosomes and more readily available nonsense suppressor tRNAs.
  • transcription and translation are often simultaneous reactions. In the absence of coupled transcription and translation in prokaryotes, mRNA stability is greatly reduced. Therefore, a prokaryotic in vitro expression system may be used which combines transcription and translation.
  • a preferred embodiment of the present invention is the use of suppressor tRNAs (especially tyrosine-inserting suppressors), which may be produced through recombinant DNA technology and/or by the partial purification of these molecules from mutant cell lines.
  • Radioactive amino acids especially S35-methionine, may be useful for monitoring in vitro translation and for following low amounts of polysomes in subsequent steps.
  • cycloheximide at a final concentration of 1 ⁇ g/ml is added to prevent the movement of the ribosomes on the mRNAs (Lynch, Meth. Enzym. 152: 248-253, 1987). This level of cycloheximide and a Mg +2 concentration of 5 mM may be use to maintain the mRNA-80S ribosome-nascent polypeptide complexes (polysomes). Other ribosome inhibitors may also be used since cycloheximide, for example, will not work on prokaryotic ribosomes. However, in the absence of GTP the polypeptide release from the ribosomes should not normally occur.
  • radioactive amino acids such as S35-methionine
  • the list of potential compounds to which the nascent peptide might bind is virtually unlimited.
  • the coupling chemistries to link these compounds to columns, matrices, filters, beads, etc., will depend to a great degree upon the nature of the compound.
  • whole cells or cellular fractions may be used to find peptides which bind to cellular components, such as receptor proteins and other membrane-bound molecules.
  • Bovine serum albumin BSA
  • gelatin casein or nonfat milk
  • other proteinaceous material is then typically added in excess to bind up any "free" surface sites.
  • an antibody is first bound to nitrocellulose by placing a solution of the antibody on a nitrocellulose disk in a microtiter dish. After absorbing the antibody to the nitrocellulose, the disk is washed by moving the nitrocellulose disk to fresh microtiter dishes containing saline. After the washes, the disk is placed in a microtiter dish containing gelatin in solution. The disk is then washed again with saline.
  • the pre-absorption step(s) may include another antibody, preferably of a similar subclass, but having different variable/hypervariable regions.
  • polysome binding may be accomplished in the presence of MgCl 2 (5 mM) and RNase inhibitors, such as heparin.
  • RNase inhibitors such as heparin.
  • nonbinding polysomes are generally removed by washings. This wash should contain MgCl 2 and perhaps gelatin, BSA, or other proteins to help reduce nonspecific binding of polysomes. If radiolabeled amino acids are used in the translations, washes (serial or flow-through) should continue until little detectable change is observed in radioactive counts bound to the substance of interest. If the amino acids are not labelled, washes should continue until at least 10 -6 dilution of the polysome solution is obtained.
  • Conditionally-binding novel peptides may be isolated after these washes by shifting the polysomes into the desired environment for nonbinding, such as higher temperature, different pH, high metal ion concentration, or low salt concentration. Those peptides (and their attached ribosome mRNA complexes) which do not bind under the second ("stringent") condition(s) will be released into the solution and represent potential conditionally-binding factors against the substance of interest.
  • conditionally-binding peptides Once immobilized, conditionally-binding peptides may be used to purify substances of interest. Alternatively, conditionally-binding peptides may serve as reagents in monitoring environmental changes.
  • the isolated (bound) polysomes may be easily disrupted by the removal of Mg +2 (by dilution or via chelating agents) or through the destruction of proteins by a number of methods (proteases, chloroform, etc.). Although dilution is the easiest method, it may not result in as thorough a disruption of the polysomes as compared to other methods.
  • the bound polysomes are placed in a solution lacking Mg +2 to liberate the mRNA; RNase inhibitors may be desirable.
  • Conditionally-binding polysomes which were released under any of the desired environments, may be treated in a similar fashion to disrupt the polysomes and release their mRNAs.
  • mRNA if a single polysome binding to the substance of interest carries a mRNA, its rare mRNA is capable of being isolated (recovered) from the entire library of mRNAs.
  • the mRNA may also be amplified by several techniques in order to facilitate isolation.
  • PCR polymerase chain reaction
  • the single cDNA may then be amplified through the use of specified primers (even the same primers as those used in cDNA synthesis).
  • the primers used for PCR may include sequences which restore the 5' and 3' regions of the original expression unit--that is, sequences which restore the promoter (e.g., the T7 polymerase recognition sequence) and 3' region are desirable.
  • sequences which restore the promoter e.g., the T7 polymerase recognition sequence
  • 3' region are desirable.
  • novel genes may be (1) cloned, (2) chemically reproduced, (3) mutated, and (4) expressed by protocols well known in the art.
  • Large-scale production of the novel polypeptide may be accomplished through recombinant DNA methods, using genetically engineered microorganisms.
  • a large variety of prokaryotic and eukaryotic expression systems exist for the in vivo synthesis of the novel binding peptide.
  • the convenient NcoI site described above or other restriction sites may be used to connect the coding region of the novel gene to a desired promoter. It will be evident to those skilled in the art that other gene splicing strategies exist as well.
  • a translation stop codon and a transcription termination sequence may be added to the 3' end of the novel gene for proper expression of the gene in a microorganism.
  • This genetically engineered sequence may then be placed on a plasmid or vector and placed within a desired host cell by transformation, transduction, infection, electroporation, microinjection, or other similar methods.
  • the novel peptide sequence may be attached to a signal sequence for possible secretion from the microorganism and/or may contain ID peptides, as herein described, for quantifying and purifying the resulting gene product.
  • the novel peptide or related sequence may be attached to other translated sequences to form a hybrid or fusion protein which is similarly expressed in a genetically engineered organism. Alternatively, large-scale in vitro transcription and translation methods may be used to produce commercial quantities of the polypeptide.
  • each cycle increases the frequency of the desired novel gene(s) by 10 4 , then three cycles may be sufficient for isolating a sequence which exists in the original library at a frequency of 10 -12 .
  • Each cycle may be completed in one to three days; and many steps of the process may be performed by automated workstations, or robots. Therefore, many cycles may be routinely accomplished for a desired binding activity within one week.
  • One aspect of the present invention does not require polysome binding for gene isolation. Instead, in vitro translation is allowed to proceed to completion, with the resultant release of the new polypeptides from the ribosome. This is accomplished by the use of nonsense codons or by the ribosomes "falling off" the end of the mRNAs.
  • the new peptides may be separated from the ribosomes and other components of the translation reaction by gel filtration and/or centrifugation and/or other means, in order to concentrate the translation products.
  • the peptide mixture is then challenged to exhibit biological or enzymatic activity--for example, the peptides are assayed for mitogenic activity by treating tissue culture cells lacking a growth factor.
  • the gene(s) which codes for this activity may be located by subdividing the original library or an RNA copy of the library and screening for activity in a subdivision. After successive subdivisions, the desired gene may be isolated to a pool containing (for example) less than 1,000 different sequences. In theory, the desired gene may be completely isolated by subdivision (to a "pool" containing just that one gene). With PCR, QB replicase or other methods (as described above), the desired sequences may be amplified to a level where in vitro transcription and translation produces a highly enriched peptide solution having the biological/enzymatic activity. At a frequency of 1 to 10 -3 , the gene of interest may be readily isolated and cloned into appropriate expression systems, using methods currently available.
  • the purified sequence codes for the activity of interest by amplifying the DNA and/or RNA so that sufficient mRNA is produced for larger-scale in vitro translation.
  • the translation products of this purified sequence should be nearly homogeneous polypeptides having the assayable activity.
  • the gene and/or the polypeptide may be sequenced by existing methods to establish the composition of the novel polypeptide.
  • the purified gene may be cloned into microorganisms for amplification and expression. Subsequently, biological/binding activities as well as sequence identity may be established for the novel gene and polypeptide.
  • hybrid proteins which have the characteristics of the novel peptide and other desirable properties.
  • One class of hybrid proteins which may be created by this technology is characterized by specific binding to cells and cytotoxic abilities. For example, a cell surface receptor-binding peptide may be joined to ricin or other toxins via DNA splicing methods. This type of hybrid protein may be used to selectively kill different cell types, including pathogens and tumor cells.
  • the gene which encodes the hybrid protein may be completely synthesized or result from splicing the appropriate gene fragments together. This gene may be expressed in a variety of expression systems.
  • a preferred embodiment of this invention is the replacement of variable and hypervariable regions of antibody and antibody-like genes by novel gene sequences which code for binding activities against substances of interest. In this manner, a much greater range of diversity is possible against antigens of interest; and the screening process may be much more efficient and time-saving than the production methods for monoclonal antibodies against the same antigens.
  • These "custom" hybrid antibody genes may be expressed in a number of organisms to produce active antibodies with new specificities or properties.
  • novel polypeptides may be considerably smaller molecules than the antibodies. Therefore, synthesis, purification, and/or manufacturing of the novel peptides may be greatly simplified and cost-effective as compared to antibodies. The smaller size may also aid in stability, formulation, and in reaching the target molecules.
  • novel polypeptides may be identifiable by (1) fusing them to a biologically active peptide which has a quantifiable activity (such as peroxidase or other enzymatic activity), (2) synthesizing them with an ID peptide, described above, to which existing antibodies are known to bind, (3) radioactively labelling them, (4) chemically adding markers, such as fluorescent dyes or metallic substances, or (5) any combination of the above.
  • a biologically active peptide which has a quantifiable activity (such as peroxidase or other enzymatic activity)
  • synthesizing them with an ID peptide, described above to which existing antibodies are known to bind
  • radioactively labelling them such as fluorescent dyes or metallic substances
  • chemically adding markers such as fluorescent dyes or metallic substances
  • novel polypeptides generated via the present invention may bind to many classes of molecules which would not elicit a strong immune response, because some molecules are not complex enough or are too similar to an organism's resident compounds to trigger antibody formation.
  • use of novel polypeptides in diagnostic binding assays may have a much greater scope than the traditional antibody-based methods.
  • novel polypeptides of the present invention may also be used therapeutically as originally isolated or as part of fusion proteins. For example, if a novel polypeptide were selected to bind a given toxin, it might also neutralize the toxin. If a new polypeptide is bound to a viral receptor site on a cell membrane or to the virus's attachment mechanism, infection of the cell may be diminished. As described earlier, fusion proteins carrying novel polypeptide recognition sequences in addition to a toxin may be used to selectively kill diseased or malignant cells. The binding of novel sequences to infected or malignant cells may trigger an immune response against the cell-peptide complex and, therefore, may be useful in the control of disease.
  • the sequences and strategies for creating a novel gene library require careful planning by those skilled in the art.
  • the 5' untranslated region of the expression unit contains an RNA polymerase site, a ribosome binding site, an initiation codon, and selected 5' untranslated sequences.
  • the polymerase binding site used in this example is the T7 promoter sequence: TAATACGACTCACTATAGGGAGA (23-mer), which is placed at the 5' end of the expression unit.
  • the ribosome binding site should include at least part of the consensus sequence for eukaryotic untranslated regions.
  • Kozak ibid., 1987
  • a selected untranslated region of 36 nucleotides is used here.
  • This untranslated region is derived from the naturally-occurring (36-base pair) upstream sequence of the adult rabbit hemoglobin (alpha-globin): A CACTTCTG G TCCAGTCCGACTGAGAAGGAACCACC ATG G, where the underlined ATG represents the start of translation at a methionine initiation codon (Baralle, Nature 267: 279-281, 1977).
  • the rabbit alpha-globin untranslated sequence is chosen because (1) it is expected to be a favorable substrate in a rabbit reticulocyte system and (2) it contains the important "motifs" of Kozak's model mRNA.
  • the alpha-globin sequence is modified in the following ways for in vitro gene expression.
  • This second change also creates a convenient GATC restriction site in the 5' untranslated region.
  • the resulting leader sequence including the ATGG of the coding region, is therefore the following: This leader sequence is placed immediately downstream from the T7 promoter.
  • the 3' region contains (1) a selected sequence for specific-primer-directed DNA synthesis, (2) a GGG-rich region which codes for a polyglycine tether that gives the nascent polypeptide spatial freedom to bind the substance of interest, and (3) convenient restriction sites whose resulting RNA secondary structure may impede the translocation of ribosomes off the mRNA.
  • the polyglycine region comprises 20 codons for glycine; most of the glycine codons are adjacent GGG triplets, which code for glycines in all reading frames. However, some of the glycine codons are GGT or GGA to keep the DNA strands in proper register.
  • the restriction sites for Bam HI (GGATCC) and NotI (GCGGCCGC) are chosen to be placed very near the 3' end of the gene; in the mRNA these sequences are expected to form hairpin loops.
  • AAAA an addition of AAAA is made at the 3' end.
  • the 3' region therefore has a general sequence of (GGG or GGT/A) 20 followed by GGATCCGCGGCCGCAAAA. A specific sequence for this region is given below.
  • the semi-random gene sequence is synthesized with known 5' and 3' ends which undergo basepairing and ligation with the fully described 5' untranslated region and 3' region segments.
  • the semi-random gene is synthesized with a 5' CACC ATG G, which may basepair with the octamer CCATGGTG on the complementary strand of the 5' untranslated region.
  • the initiation (first) codon, ATG is necessary for translation of the semi-random sequences.
  • the subsequent G is the first position of the second codon and is constant to preserve the NcoI site at the front end of the gene.
  • the rest of this second codon and the next 28 codons are synthesized following the rules outlined earlier for reducing nonsense triplets.
  • Sequences I and II are mixed in equimolar amounts in standard TE Buffer and heated at 65°C for 5-10 min.
  • the complementary sequences (which comprise the 5' untranslated region) are allowed to anneal at 50°-60°C for one hour or longer, are allowed to cool slowly to room temperature, and are thereafter stored at 0°-4°C.
  • Sequences IV and V are likewise treated to form the double-stranded 3' region. These duplexes each have an eight-base, single-stranded overhanging sequence which is complementary to the known ends of Sequence III.
  • 10 ⁇ g of completely double-stranded DNA molecules has a sequence diversity of 4 x 10 13 .
  • This library may then be transcribed with T7 RNA Polymerase to yield translatable mRNAs. However, with each transcription, the DNA library is consumed, unless DNA copies are made.
  • 100 ng aliquots are each distributed to 500- ⁇ l tubes for PCR amplification in 200- ⁇ l reactions. According to PCR Technology, pp. 18-19 (Erlich, ibid., 1989), each 200- ⁇ l PCR reaction yields about 5.2 ⁇ g of DNA--or an approximately 50-fold duplication of DNA in each aliquot. The aliquots are pooled.
  • the pooled sample contains on the average 50 copies of each semi-random sequence and therefore may be used repeatedly (50 times, for example) without a large loss of diversity for each translation with T7 RNA Polymerase. If the library is to be replicated with PCR, then the Klenow filling and ligation steps, described above, may be unnecessary, since the Taq polymerase is capable of filling in the gap and nick-translating DNA (D. H. Gelfand, PCR Workshop, Society of Industrial Microbiology Meeting, Seattle, Wash., 1989). After nick translation, the gene is double-stranded and able to be PCR amplified.
  • oligonucleotide primers for PCR amplification of the DNA library are listed above in sequences I and VI. Generally, oligonucleotides of 25-30 bases are used for PCR amplification; however, longer primers may be used. It is important that the primers do not share significant homologies or complementary 3' ends. Sequences I and VI have noncomplementary ends and no obvious regions of extensive homology.
  • sequence VI is used as the primer for first-strand cDNA synthesis .
  • Sequence I is used as the primer for second-strand synthesis and restores the T7 promoter to the cDNA. In this way, later rounds of translation are possible on the selected novel gene sequences. PCR amplification may be necessary if the resulting cDNAs are relatively rare.
  • the DNA library (or a representative aliquot of those sequences) described in Example One is transcribed with T7 RNA polymerase. 2.5 ⁇ g of this DNA codes for nearly 10 13 different polypeptides.
  • the DNA is capped during transcription with Stratagene's mCAPTM Kit, according to the manufacturer's specifications. Approximately 5-10 ⁇ g of mRNA is expected. Generally, with T7 RNA polymerase, nearly 10 times this level of RNA is synthesized; however, the conditions for the capping reaction limit mRNA production in this case.
  • the DNA is removed with DNase I, provided in the kit.
  • the capped mRNA is phenol/chloroform extracted and precipitated with ethanol. The RNA is resuspended in 10 ⁇ l of TE and stored at 0°-4°C.
  • the capped mRNA is translated with Boehringer Mannheim Biochemical's rabbit reticulocyte kit, with all 20 amino acids at 312.5 ⁇ mol/l each.
  • Capped mRNA from Example 2 is added to each reaction at 0.5 ⁇ g per reaction and is treated according to the manufacturer's protocol. After around 60 minutes at 30°C, cycloheximide is added to a final concentration of 1 ⁇ g/ml. MgCl 2 is adjusted to 5 mM, and heparin is added to 0.2 mg/ml.
  • the reactions are pooled and submitted to a discontinuous sucrose gradient, according to Lynch (ibid., 1987).
  • the polysomes may be frozen at -70°C or used directly.
  • Antibodies may be used to select for novel binding peptides. Peptides which bind to the hypervariable/variable regions of the antibodies ("anti-id peptides") may behave like the original epitopes which were used as immunogens. Because the novel anti-id peptides may mimic the original epitopes, these peptides may be useful as vaccines and/or may demonstrate biological activities, in much the same way that anti-id antibodies have been shown to have biological (sometimes catalytic) activities.
  • antibodies examples include anti-fibronectin, anti-nerve growth factor, anti-CD4, and anti-tumor necrosis factor, which are all available from Boehringer Mannheim Biochemicals.
  • antibodies to receptor molecules, growth factors, surface antigens, and biologically active peptides, as well as neutralizing antibodies to toxins and diseases are good candidates for which to isolate anti-id binding peptides that may have agonist or antagonist properties or serve as vaccines.
  • the antibodies are affixed to Immobilon" PVDF (polyvinylidene difluoride) membrane from Millipore Corporation, according to Pluskal et al. ( BioTechniques 4 :272-283, 1986).
  • Immobilon PVDF (polyvinylidene difluoride) membrane from Millipore Corporation, according to Pluskal et al. ( BioTechniques 4 :272-283, 1986).
  • anti-fibronectin antibody from clone 3E3, Boehringer Mannheim Biochemicals
  • Immobilon PVDF is reported to bind 172 ⁇ g/cm 2 of IgG.
  • 1 ⁇ g of anti-fibronectin IgG 1 in saline buffer is absorbed onto the PVDF square by incubating at room temperature for at least two hours.
  • the PVDF is then washed with the Saline Buffer twice.
  • the membrane is next incubated with a "blocking solution,” containing 5% (w/v) gelatin in saline buffer for at least two hours at room temperature, so that the gelatin is absorbed into unoccupied sites of the PVDF.
  • the membrane is then washed twice with 0.1% gelatin in saline buffer.
  • a similar treatment is done with 10 ⁇ g anti-keratin antibody (from clone AE1, Boehringer Mannheim Biochemicals), which is the control IgG 1 as described below.
  • Polysomes with nascent semi-random peptides are incubated in 1-ml reactions, each containing PS Buffer (0.9% NaCl, 10 mM Tris pH 7.4, 1% gelatin, 15 mM MgCl 2 , 0.2 mg/ml heparin, and 1 ⁇ g/ml cycloheximide) and a PVDF square with 10 ⁇ g anti-keratin IgG 1 , described in Example 4.
  • This pre-absorption step is done at 0°-4°C with gentle agitation for four hours to select out nonspecific binding of polysomes to gelatin and IgG 1 .
  • the anti-keratin PVDF square is removed with jewelers' forceps and is replaced with the anti-fibronectin PVDF square.
  • the mixture is incubated for four more hours under the same conditions to allow specific polysome binding to the variable/hypervariable region of the anti-fibronectin antibody.
  • the anti-fibronectin PVDF square is removed and washed three times by transferring it serially to fresh PS buffer.
  • the PVDF membrane which holds the washed antibody-bound polysomes, is transferred to a tube containing 100 ⁇ l of 0.1 mM EDTA and is gently shaken at room temperature for 5-10 minutes to disrupt the polysomes and liberate mRNA.
  • the PVDF is removed, placed in a fresh tube of 0.1 mM EDTA, and stored at 0°-4°C overnight or longer (as a back-up).
  • the released mRNA from the first EDTA treatment is reverse transcribed; and the resulting cDNA is amplified, according to PCR Technology (ibid., 1989), p. 91, with slight modification.
  • a sequence complementary with the known 3' region (such as Sequence VI listed earlier as the downstream primer) is used for both cDNA synthesis and PCR reactions.
  • the reverse transcriptase step is done in 100 ⁇ l of PCR buffer with the appropriate relative amounts of the other reagents (instead of 20- ⁇ l reaction). After the reverse transcriptase reaction, the mixture is split into 20 ⁇ l aliquots; and each aliquot is amplified as described in PCR Technology, using Sequence I or a similar DNA upstream primer. After PCR amplification, the five aliquots are pooled, phenol/chloroform extracted, and ethanol precipitated. This cDNA is then resuspended in TE and stored at 0°-4°C.
  • the selected DNA is transcribed with T7 RNA polymerase and translated in a reticulocyte system, as previously described.
  • the desired sequences are greatly amplified compared to the original DNA library.
  • desirable novel genes are concentrated to a level where conventional cloning and expression methods are practical.
  • a single novel gene(s) may be isolated, amplified, transcribed, and translated to demonstrate specific binding capability of the gene product(s). Once binding has been demonstrated, the isolated gene(s) and polypeptide(s) may be sequenced for identification.
  • the amplified pools of recovered cDNA are assayed for the presence of the genes.
  • ID sequences have been intentionally included to be coexpressed with the semi-random DNA sequences
  • ELISAs or other immunological assays for the known part of the peptide are used to detect the binding of the novel portion of the peptide to the substance of interest.
  • competition assays for the peptides are carried out. Competition assays, including competition ELISA tests, are used to monitor for the presence of binding sequences within the various cDNA pools generated by the present invention.
  • One example is the screening of the cDNA pools for genes which encode peptides which bind anti-Pseudomonas exotoxin (anti-PE) antibody.
  • anti-PE anti-Pseudomonas exotoxin
  • different aliquots of the resultant cDNA pool were each transcribed in a 200- ⁇ l reaction with T7 RNA polymerase (30 units) under standard conditions, starting with approximately 200 ng of DNA.
  • the mRNA products were phenol/chloroform/isoamyl alcohol extracted and precipitated with sodium acetate and ethanol at -20°.
  • the precipitates were each centrifuged and resuspended in 16 ⁇ l of distilled water which had been treated with diethylpyrocarbonate to remove nucleases.
  • RNA samples were heated to 65° for five minutes and then placed on ice.
  • the RNAs were translated with a wheat germ kit (Boerhinger Mannheim Biochemicals) according to the manufacturer's recommendations.
  • Each RNA sample was expressed in a 50 ⁇ l translation reaction with 25 microcuries of 35S-methionine and 0.5 ⁇ l of RNase inhibitor. The reactions were run for 15-60 minutes at 30°. At the end of the translation, the samples were each equally divided: one half was used to bind the substance of interest without competing substrate, while the other half was used to bind the substance of interest in the presence of excess competing substrate. In this case, the substance of interest was anti-PE antibody.
  • the competing substrate was a 14-amino acid peptide (PE peptide), which is derived from the toxin protein sequence and known to bind the antibody.
  • PE peptide 14-amino acid peptide
  • the PE peptide sequence is Val-Glu-Arg-Leu-Leu-Gln-Ala-His-Arg-Gln-Leu-Glu-Glu-Arg. See Wozniak, et al., Proc. Natl. Acad. Sci., 85: 8880-8884 (1988).
  • each disk was serially washed eight times in 200 ⁇ l of fresh TSM at 0°, with a 10 minute incubation for each wash.
  • the bound radioactivity for each disk was measured in a liquid scintillation counter with a 1 ml cocktail of Ecoscint.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Analytical Chemistry (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

A method for the cell-free synthesis and isolation of novel genes and polypeptides is provided. Within one embodiment, an expression unit is constructed onto which semi-random nucleotide sequences are attached. The semi-random nucleotide sequences are first transcribed to produce RNA, and then translated under conditions such that polysomes are produced. Polysomes which bind to a substance of interest are then isolated and disrupted; and the released mRNA is recovered. The mRNA is used to construct cDNA which is expressed to produce novel polypeptides.

Description

Technical Field
The present invention generally relates to the synthesis and isolation of novel genes and polypeptides in vitro and, more specifically, to methods of generating and expressing semi-random DNA or RNA sequences, isolating novel genes from those sequences, and using those genes to create novel polypeptides.
BACKGROUND OF THE INVENTION
The isolation of novel genes and polypeptides from semi-random sequences is currently limited by the need to screen a large, genetically diverse population of cells in order to obtain the sequence(s) of interest. For example, a polypeptide string of 10 amino acids has 2010 or approximately 1013 possible permutations. If 10 of these permutations had a desirable characteristic (such as the ability to bind a specific antigen), then a population of 1012 would have to be screened for the expectation of finding one desirable novel gene. Through the use of conventional methods (expressing novel genes via microorganisms), the screening of a large number of new sequences for a specific property is virtually unfeasible, unless the novel gene provides the organism with a distinct growth or survival advantage. Indeed, under the current state of the art, the 1012 independently transformed microorganisms would have to be screened individually to locate that one desirable novel gene.
Within present screening procedures for detecting novel gene products which are localized within cells, colonies derived from each transformed cell must be treated to break open the cells. Typically 1000-2000 bacterial colonies per standard petri dish are lysed (e.g., by chloroform) for the screening procedure. Thus, to examine 1012 transformed organisms, 500,000 to 1 billion petri dishes would be necessary. In addition, 10,000 to 100,000 liters of logarithmically dividing cells may be necessary for producing the large numbers of transformable cells.
Alternatively, where a gene product is secreted and attached to the outside of a cell, it may be detected by its ability to bind a fluorescent compound or other marker. In these cases, cell sorters may be used to screen for the synthesis of a novel desirable polypeptide. However, even at a flow rate of 5,000 cells per second, it would take a cell sorter over 60 years to screen 1012 cells. Thus, present day screening methods which are both extremely costly and time-consuming, effectively prohibit the isolation of novel genes and polypeptide from semi-random sequences.
In addition to the methods briefly discussed above, Fields and Song (Nature 340:245-246, 1989) proposed a method for selectably obtaining polypeptides which specifically bind to other polypeptides, using the domains of the yeast GAL4 gene. However, this system has serious limitations. First, only polypeptide-polypeptide binding may be selected; polypeptide-nonpolypeptide interactions are excluded. Second, both the known and novel binding polypeptides have to be expressed in yeast at reasonably high levels and in "native" conformations for the method to have commercial applicability. Third, glycosylated polypeptides or polypeptides that have special modifications may also be excluded by this method. Fourth, it is not clear whether random or semi-random sequences can work, given that they used known polypeptides whose physical interactions were well-established and yet showed only 4.5% of the control GAL4 activity. Fifth, Fields and Song used very large sequences: 633 amino acids of the SNF1 protein and 322 amino acids of the SNF4 protein, which have evolved secondary structures that interact with each other. Sixth, using their method for semi-random sequences of even 1010 diversity obviates the need for extremely large amounts of DNA, modifying enzymes, and competent yeast cells.
Contrary to previously disclosed methods, the present invention describes a method for cell-free screening of novel genes and polypeptides. This method avoids the problems associated with large numbers of transformed organisms as well as the limitations of the method disclosed by Fields and Song, and may be completed within a few weeks. Therefore, the methodology allows a substantial time and monetary saving in the isolation of novel gene products.
SUMMARY OF THE INVENTION
Briefly stated, the present invention relates to methods for synthesizing, screening, and selecting high numbers of novel genes and polypeptides.
According to a first aspect of the present invention there is provided a method for isolating a nucleotide sequence which encodes a polypeptide of interest, comprising transcribing or replicating an in vitro expression unit which comprises a 5' untranslated region containing a RNA polymerase binding sequence, a ribosome binding sequence, a translation initiation signal, and one or more semi-random nucleotide sequences to produce a mRNA library; translating the mRNA library under conditions which maintain polysomes having polypeptide chains attached thereto; contacting the polysomes to a substance of interest that is capable of specifically reacting with the polypeptide of interest and isolating mRNA from polysomes that specifically react with the substance of interest.
In one embodiment of the method described above, the process may be repeated on mRNA that has been enriched for desirable sequences by amplifying the RNA or respective cDNA. Subsequently, this amplified subset of genes may be cycled through the various steps outlined above to further enrich for desirable novel genes until desirable sequences represent a significant (>10-3) fraction of the truncated population. In principle, the method may be repeated until the population of genes is nearly homogeneous.
Within a second aspect of the present invention, there is provided a method for producing novel polypeptides by expressing the nucleotide sequence isolated in the method of the first aspect comprising, subsequent to the contacting step of the first aspect, isolating said polysomes that bind to said substance of interest; disrupting said isolated polysomes to release mRNA; recovering said mRNA; constructing cDNA from said recovered mRNA; and expressing said cDNA to produce novel polypeptides.
In a third aspect of the present invention, there is provided a method for producing novel polypeptides by expressing the nucleotide sequence isolated in the method of the first aspect wherein, during said translating step, the conditions optionally permit polypeptide chains to be released from the polysomes, and further comprising isolating the mRNA encoding the polypeptides identified in the contacting step by subdividing, repeating the steps of transcribing, translating, contacting and isolating by subdividing, such that the gene of interest is isolated; constructing cDNA from said isolated gene; and expressing said cDNA to produce novel polypeptides.
The expression unit described above comprises an RNA polymerase binding sequence, a ribosome binding site, and a translation initiation signal. The expression unit may further comprise a translation enhancer or "activator" sequences, a 3' tail of a selected sequence and appropriate restriction sites. The semi-random DNA sequences may be generated by mechanically, chemically, or enzymatically fragmenting naturally-occurring DNA, by chemically synthesizing the DNA, or by polymerizing the DNA directly onto the expression unit. The substance of interest may be a surface antigen, receptor protein, toxin, organic polymer, active site of a protein molecule, metabolite, antibody, metal, hormone, or other compound.
These and other aspects will become evident upon reference to the following detailed description.
DESCRIPTION OF THE PREFERRED EMBODIMENT
The present invention is directed to the isolation of novel genes and polypeptides. These novel genes may have virtually infinite diversity and may code for new polypeptides with commercially important properties, such as novel catalytic activities or the ability to bind selectively to specific substances. Novel genes may be constructed which comprise open reading frames from existing genes or from semi-random nucleotide sequences of chemically synthesized DNA. They may be expressed in a wide variety of organisms using existing promoters, enhancers, initiation codons, plasmids, ribosomal binding sites, and/or terminators. In some cases, it may be advantageous to express the novel genes in vitro, as part of a large-scale production process.
As noted above, the present invention describes a multistep process for constructing and isolating novel genes and gene fragments which encode novel polypeptides with specific binding and/or biological activities. Within a preferred embodiment, the process comprises the following steps:
  • 1. An expression unit is constructed which contains a RNA polymerase binding sequence (i.e., a promoter or a RNA-directed RNA polymerase initiation site), a ribosome binding site, and a translation initiation signal. The expression unit may also contain convenient restriction sites, translation enhancer or "activator" sequences, and a 3' tail of a selected sequence.
  • 2. Semi-random DNA or RNA sequences are then generated by mechanically, chemically, or enzymatically fragmenting naturally-occurring DNA, RNA, or cDNA sequences, or by chemically synthesizing the nucleotides. The semi-random DNA or RNA sequences are then inserted into the expression unit. Alternatively, the semi-random sequences can be polymerized directly onto the expression unit. A library of 1012 or greater different sequences may then be created.
  • 3. The novel genes are then transcribed in vitro to produce a pool of RNA copies of the original DNA library. If an RNA-directed RNA polymerase sequence is included, then these replicases may be used to amplify the RNA.
  • 4. The RNA (mRNA) is translated in vitro to produce polysomes. Conditions for maintaining the "polysomes" (RNA-ribosome-nascent polypeptide complexes) are used to keep the desired polypeptide and mRNA together.
  • 5. The polysomes are then allowed to bind to substances of interest, such as surface antigens, receptor proteins, toxins, organic polymers, antibodies, metabolites, hormones, and active sites of protein molecules, or to display biological activity.
  • 6. Polysomes binding to the substance(s) of interest are substantially enriched by the removal of the unbound polysomes. Serial or flow-through washes under conditions which maintain the polysome complexes substantially increase the frequencies of the desired mRNAs, which remain attached to the substances of interest through the polysome structure.
  • 7. The bound/active polysomes are then disrupted to release the mRNAs from the polysome complex.
  • 8. The rare mRNAs are then recovered by making cDNA copies or by direct amplification of the RNA with RNA-directed RNA polymerases. The amplification of the cDNA with DNA polymerase and/or reverse transcriptase reactions may allow greater ease in recovering these low abundance messages.
  • 9. The resulting cDNAs are then expressed to produce polypeptides.
  • In most instances, repetition of steps 3-8 is preferable to further increase the frequency of specific binding proteins above a background of nonspecific binding of polysomes.
    The isolated, purified novel gene(s) produced by the methods described herein are capable of generating a variety of polypeptide(s) of interest using standard expression techniques, as positive proof that the gene codes for the desired product. In addition, DNA and/or polypeptide sequencing by conventional methods may be used to identify the composition of the novel polypeptide.
    Once the polypeptide encoded by the novel gene has been isolated and identified, large-scale production of the novel polypeptide(s) may be accomplished by chemical synthesis (if the amino acid sequence is relatively short) or through recombinant DNA methods, using genetically engineered microorganisms. Alternatively, large-scale in vitro transcription and/or translation methods may be used to produce commercial quantities of the polypeptide.
    The DNA sequence coding for the selected polypeptide may also be incorporated into larger genes (i.e., such as into the hypervariable regions of antibody genes) to create hybrid proteins with the specific binding and/or biological activities of the originally isolated novel polypeptides, in addition to other binding and biological activities.
    I. THE EXPRESSION UNIT
    The expression unit comprises a 5' untranslated region and may additionally comprise a 3' region. The 5' untranslated region of the expression unit contains a promoter or RNA polymerase binding sequence, a ribosome binding sequence, and a translation initiation signal. The 5' untranslated region ("head") may also contain convenient restriction sites and a translation enhancer or "activator" sequence(s). The 3' region may contain convenient restriction sites and a 3' tail of a selected sequence. The expression unit may be chemically synthesized by protocols well known to those skilled in the art. Alternatively, these elements may be incorporated into one or more plasmids, amplified in microorganisms, purified by standard procedures, and cut into appropriate fragments with restriction enzymes before assembly into the expression unit.
    The 5' untranslated region contains a promoter or RNA polymerase binding sequence. High-efficiency promoters, such as those for the T7, T3, or SP6 RNA polymerase, are preferred in this invention for the following reasons. Such promoters are short DNA sequences of known composition, are extremely specific for their relative polymerases, and are highly active, allowing for greater than 50 rounds of transcription per DNA template. In addition, T7, T3, and SP6 polymerases are commercially available from many sources and are components of well-characterized transcription kits. For the T7 promoter, the consensus sequence is TAATACGACTCACTATAGGGAGA (23 base pairs). Although this sequence is described in conjunction with a preferred embodiment of this invention, it will be evident that related DNA sequences may be used which will function for T7 RNA polymerase, and other sequences will be appropriate for other RNA polymerases. Within certain embodiments, it may be desirable to utilize two promoters, such as both the T7 and SP6 promoters.
    Positioned downstream of or within the promoter region is a DNA sequence which codes for a ribosomal binding site. This ribosome binding site may be specific for prokaryotic ribosomal complexes (including ribosomal RNAs) if a prokaryotic translation procedure is used. However, a preferred embodiment of this invention uses a eukaryotic sequence and an in vitro eukaryotic translation system, such as the rabbit reticulocyte system (Krawetz et al., Can. J. Biochem. Cell. Biol. 61:274-286, 1983; Merrick, Meth. Enzymol. 101:38, 1983). A consensus translation initiation sequence, GCCGCCACCATGG, as well as other functionally related sequences have been established for vertebrate mRNAs (Kozak, Nucleic Acids Res. 15:8125-8148, 1987). This sequence or related sequences may be used in the novel gene construction to direct protein synthesis in vitro. The ATG triplet in this initiation sequence is the translation initiation codon for methionine; in vitro protein synthesis is expected to begin at this point.
    Between the promoter and translation initiation site, it may be desirable to place other known sequences, such as translation enhancer or "activator" sequences. For example, Jobling et al. (Nucleic Acids Res. 16:4483-4498, 1988) showed that the untranslated "leader sequences" from tobacco mosaic virus "stimulated translation significantly" in SP6-generated mRNAs. They also reported that the 36-nucleotide 5' untranslated region of alfalfa mosaic virus RNA 4 increases the translational efficiency of barley amylase and human interleukin mRNAs (Jobling and Gehrke, Nature 325:622-625, 1987). Black beetle virus (Nodavirus) RNA 2 (Friesen and Rueckert, J. Virol. 37:876-886, 1981), turnip mosaic virus, and brome mosaic virus coat protein mRNAs (Zagorski et al., Biochimie 65:127-133, 1983) also translate at high efficiencies. In contrast, certain untranslated leaders severely reduce the expression of the SP6 RNAs (Jobling et al., ibid., 1988).
    Appropriate restriction sites may also be included in the expression unit to assist in future genetic engineering. For example, the sextuplet, CCATGG, is the recognition sequence for the restriction endonuclease, NcoI. A NcoI "cutting site" positioned downstream of the ribosomal binding site is a convenient splice point for subsequent genetic engineering. Hence, after purification of a desired novel gene, the expression unit may be spliced from the novel gene at this site, and another promoter may be attached for expression in vivo and large-scale production of the novel polypeptide. The NcoI site may also be used as a convenient cloning site for the construction of hybrid proteins, where two different polypeptide domains are brought together and expressed as a single protein.
    In addition, it is most likely advantageous to include in the 5' untranslated region a DNA sequence with at least one restriction endonuclease site for subsequently cloning the novel gene into plasmids. The octameric sequence, GCGGCCGGC, is recognized by NotI nuclease and is particularly useful because it would rarely fall within the novel coding region of the gene (NotI is expected to cut totally random DNA once every 65,536 base pairs). Other restriction sites may also be used; the expected frequency of cutting the novel coding region is dependent upon the nucleotide composition or the DNA source of the coding region. It should be noted that certain palindromic sequences may interfere with translation; however, some sequences may also enhance the rate of translation.
    The expression unit may also comprise a 3' region. It is desirable to construct known 3' regions (tails) with palindromic sequences for at least two reasons. First, 3' restriction sites would be convenient for any later genetic engineering of the polypeptide coding region. For example, if NotI sites were located in both the 5' and 3' regions, a desirable polypeptide coding sequence could be cut out with NotI "sticky ends" for further cloning. Second, palindromes may cause secondary structures which impede translocation, thus, palindromes in the 3' region may slow down the movement of ribosomes during translation. This second property may be desirable for preventing ribosomes from "falling off" the mRNA and thereby enhancing the number of polysomes in the in vitro translation step. The 3' region may also contain a poly-A or other polynucleotide stretch for later purification of the mRNA from other components in the in vitro translation reaction by hybridization to a complementary homopolymeric sequence.
    In addition, other nonrandom sequences may be incorporated into the expression unit. Within one embodiment, the expressed polypeptides contain both nonrandom and semi-random amino acid sequences. The nonrandom component of the coding region is synthesized and produced with the nonrandom 5' untranslated region and/or with the 3' region. This nonrandom coding sequence specifies a string of amino acids (an identification or "ID" peptide) that is conserved among the billions of novel polypeptides. The ID peptide would be useful for quantifying the amount of novel polypeptide and for purification of the novel polypeptide (given that an antibody against the ID peptide is available or can be produced). One example is the 11 amino acid Substance P, which can be attached as a fusion peptide to other polypeptides. Anti-Substance P antibodies are commercially available for detecting and quantifying fusion proteins containing Substance P. Another example is the eight amino acid marker peptide, "Flag" (Hopp et al., Bio/Technology 6:1204-1210, 1988).
    Amino-terminal ID peptides have at least two advantages over carboxy-terminal ID peptides. First, it is easier to make gene constructions which maintain the proper reading frame of the N-terminal ID, because long stretches of semi-random DNA or RNA will tend to end in all three reading frames for a C-terminal ID. Second, the N-terminal ID may be designed to function as a signal peptide in a transformed organism, allowing for the possible secretion of the novel polypeptide during large-scale production.
    Nevertheless, C-terminal ID polypeptides may also be used. One preferable C-terminal polypeptide is polyglycine, which is encoded by poly-dG and is read Gly-Gly-Gly, etc., regardless of the reading frame of the semi-random sequences. The polyglycine 3' end of the polypeptide may act as a noninterfering tether of the nascent peptide and allow the semi-random sequences greater access to bind molecules of interest. In addition, the poly-dG sequence may be used for priming second strand synthesis of the cDNA and may be useful for purification of the RNA or DNA with polyC or poly-dC. Other repetitive sequences, such as GGGCGGGC..., may be used to code for a recognizable peptide sequence which is expressed in all reading frames. A preferable form of the ID peptide is one which may be cleaved from the novel polypeptide by simple chemical or enzymatic means.
    In addition to the DNA expression unit, an RNA expression unit may be constructed for semi-random polypeptide synthesis. One possible advantage of the RNA expression unit is that the recovery of the polysomal mRNA does not have to go through an initial cDNA stage. Instead, the mRNA with the desired sequences may be amplified with an RNA-directed RNA polymerase, such as that of QB (Q Beta) replicase (Haruna and Spiegelman, Proc. Nat. Acad. Sci. 54:579-587, 1965). This enzyme can make one billion copies of recombinant RNA in 30 minutes (Lizardi et al., Bio/Technology 6:1197-1202, 1988). One suitable cloning strategy for amplification of recombinant RNA is detailed in Lizardi et al. (ibid., 1988). For purposes of the present invention, other elements, such as restriction sites, enhancers, and ID sequences, may be added to the DNA plasmids which give rise to the QB RNA templates. Semi-random coding sequences may be inserted on these plasmids by standard DNA methodologies. When the QB replicase template is transcribed (for example, by T7 RNA polymerase), an RNA library capable of in vitro replication may be created which contains the semi-random gene sequences. Alternatively, a similar RNA expression unit may be constructed by chemically synthesizing the appropriate RNA molecules and assembling them via an RNA ligase, such as the T4 RNA ligase (commercially available), which links together single-strand RNA and/or single-strand DNA.
    II. SEMI-RANDOM NUCLEOTIDE SEQUENCES
    Semi-random sequences of DNA or RNA are attached to the expression unit. Since the RNA expression units and semi-random sequences may be generated from a DNA template or constructed from chemically synthesized RNA or mRNA fragments in much the same manner as DNA expression units, the following description merely describes the process for semi-random DNA attachment to the expression unit. Those skilled in the art will readily be able to construct the RNA-equivalent of the expression units attached to semi-random polynucleotides.
    Semi-random DNA may be generated by at least three methods. First, naturally-occurring DNAs from virtually any living source may be mechanically, chemically, or enzymatically fragmented and attached to the 5' untranslated region with DNA ligase. Mixtures of fragments from different DNA sources may be used. The end result may be the selectable expression of an active "open reading frame"--a portion (fragment) of a protein that has no "nonsense" (or "stop") codon, unless the activity resides in the extreme C-terminus of the molecule. In one embodiment of this invention, a gene coding for a known function may be fragmented; the resulting pieces are ligated to the 5' untranslated region and later screened for the expression of activity in the polysome assay. By examining the smallest gene fragment which provides biological activity, an analysis of protein domains may be made. Gene fragment analysis may be useful for creating small biologically active peptides and hybrid therapeutic proteins and may be beneficial for drug delivery, if smaller size assists the peptide in reaching the target site.
    In another embodiment of the present invention, the "fragmented" DNAs may be semi-randomly sized cDNA molecules from a cDNA library. By expressing cDNAs in vitro and using polysome selection, a very rare partial or perhaps even full-sized gene may be isolated through binding the polysome to antibody, receptor protein, or other diagnostic molecule. The cell-free expression of cDNA "fragments" as herein described may be orders of magnitude more sensitive than previously described methods in locating desirable cDNA clones.
    A second method for generating semi-random DNA is to chemically synthesize the DNA. For example, relatively long DNA molecules of approximately 100 nucleotides may be synthesized with mixtures of nucleotides at each position. However, a statistical problem of nonsense codons becomes apparent with chemically synthesized DNA. For the gene fragments and cDNA strategies described above, an active, open reading frame is located from within existing protein sequences. "Open reading frame" implies that no stop codon exists and often indicates a sequence from within a protein coding region.
    However, it should be noted that chemically synthesized DNA having enough diversity to code for all 20 common amino acids at all positions may not necessarily have open reading frames. The stop codons--TAA, TAG, and TGA--represent three of the 64 possible DNA triplets. For completely random DNA, with the equal likelihood of any of the four nucleotides in each position, the probability of a nonsense codon is therefore 3/64 = 4.6875%. For a random DNA stretch coding for a string of 30 amino acids, the probability of at least one stop codon within that string is about 76%. Stop codons cause termination of translation and release of the nascent polypeptide from the ribosome complex. Therefore, strategies to reduce the frequencies of nonsense codons and to bypass the usual result of nonsense codons during protein translation are preferable, and discussed below.
    More specifically, the A, T, C, and G base composition may be manipulated to favor certain codons and in particular to reduce the likelihood of nonsense codons. In the extreme case, the third position of each triplet codon may be synthesized with only C and T to theoretically avoid nonsense codons. However, in this case not all 20 amino acids are encoded. Lim and Sauer (Nature 339:31-36, 1989) have used an equal mixture of all four bases in the first two codon positions and an equal mixture of C and G at the third codon position in synthesizing new regions of lambda repressor. This combination allows for any of all 20 amino acids at each codon and reduces the frequency of nonsense triplets to 1/32 = 3.125%. However, in a string of 30 amino acids the likelihood of at least one TAG stop codon is about 61%.
    In a preferred embodiment of this invention, unequal mixtures of the bases are used in all three codon positions to reduce the frequency of stop codons, while still allowing a high frequency of all 20 amino acids at all codons. In the first codon position equal molar amounts of C, A, and G are used, but only half that amount of T is used. In the second codon position the amount of A is reduced to half of the level of the other three bases. In the third codon position only G and C or G and T are used, and in equal molar amounts. The result of this strategy is a greater than 79% probability that no stop codons will be present in a string of 30 amino acids. The proportions of the individual amino acids are slightly distorted in this case relative to a totally random DNA strategy. However, only tyrosine will be represented at less than half of the expected frequencies compared to the random situation.
    To further overcome the presence of nonsense codons when using chemically synthesized DNA, it is preferred that nonsense suppressing tRNAs be used in the in vitro translation steps. In particular, since the strategy described above eliminates all but the TAG stop triplet, and tyrosine codons are underrepresented as the result of unequal mixtures of bases at each codon position, a nonsense suppressor which recognizes TAG (actually UAG in the mRNA) and inserts tyrosine into the growing polypeptide chain is most desirable. Such tyrosine-inserting nonsense suppressors may be generated by changing the anticodon region of a tyrosyl-tRNA in such a manner that the tyrosyl-tRNA now "reads" UAG instead of the normal UAU and UAC tyrosine codons in mRNA. Normal tyrosyl-tRNAs will also be included in the translation step to read the tyrosine codons. Nonsense suppressors can also be made for the other two nonsense codons. As an example, tryptophane- or leucine-inserting suppressors of the UGA stop codon have been well characterized--as have many other nonsense suppressors. The nucleotide sequences of many nonsense suppressors are known; and, therefore, the construction of such molecules would be evident to those skilled in the art.
    Nonsense suppressors of mammalian translation systems are known (Burke and Mogg, Nucleic Acids Res. 13:1317-1326, 1985; Capone et al., EMBO J. 4:213-221, 1985; Diamond et al., Cell 25:497-506, 1981; Hudziak et al., Cell 31:137-146, 1982; Laski et al., EMBO J. 3:2445-2452, 1984). Additionally, different investigators have shown that the "reading" of nonsense codons in eukaryotic in vitro translation systems is possible with the use of suppressor tRNAs, including the tyrosine-inserting UAG suppressor tRNA from yeast (Capecchi et al., Cell 6:269-277, 1975; Gesteland et al., Cell 7:381-390, 1976). Readthrough of the UAG stop codon by such yeast suppressors has been reported as high as 70% in vitro (Pelham, Nature 272:469-471, 1978). Geller and Rich (Nature 283:41-46, 1980) have successfully suppressed nonsense codons in reticulocyte systems with yeast suppressor tRNAs and with bacterial suppressor tRNAs and tRNA synthetase. Therefore, the use of tRNA suppressors in the present invention to reduce premature release of polypeptides from the ribosomes during the translation step is well within the state of the art. Furthermore, both Pelham (ibid., 1978) and Geller and Rich (ibid., 1980) describe high levels of naturally-occurring nonsense suppression in eukaryotic translation systems. In particular, Pelham shows that a particular UAG codon in tobacco mosaic virus may be "read" (suppressed) nearly 40% of the time by "supraoptimal concentrations of Mg+2", or a reported 2.1 mM MgCl2. This level of magnesium ion or higher may therefore be used advantageously within the present invention to increase the readthrough of nonsense codons and to thereby reduce the problem of translation termination of longer semi-random nucleotide sequences.
    In generating the semi-random DNA by chemical means, different mixtures of bases at selected codon positions may be used to strongly bias in favor of or against a particular amino acid. For example, the elimination of G at position three in a codon prevents methionine and tryptophan from being included in the peptide. As another example, a nucleotide mixture which is biased toward a high-cysteine content may be desirable for producing short peptides with internal disulfide bonds for structural rigidity. Such rigid peptides may bind other molecules more tightly.
    Second-strand synthesis of these artificial nucleotide sequences may be accomplished by "random priming" and extension with DNA polymerase and/or by including a poly-dX tail from which to prime with poly-dX'. Other methods, such as the use of terminal palindromes that create "hairpin loops" for self-priming, may be used for second strand synthesis. 100 µg of double-stranded DNA of 100 nucleotides contains about 1015 molecules. If the semi-random synthesis strategy is used, the expectation is that each of these molecules codes for a different polypeptide. Therefore, a very large diversity in coding potential exists within laboratory bench-scale amounts of DNA. Such a synthetic DNA molecule of 100 nucleotides is merely provided for purposes of illustration; longer sequences may also be synthesized. In addition, shorter synthetic molecules may be generated and ligated together to make semi-random sequences of any given length. Shorter molecules are expected to preserve the reading frame of the synthetic DNA better than longer molecules, because each addition of chemically synthesized base is not 100%. Therefore, more nonsense codons may be avoided by the use of shorter artificial DNA molecules. T4 RNA ligase or other means may be used to link together the short single-stranded DNAs.
    A third method for generating semi-random DNA is to polymerize the molecules directly onto the 3' end of the 5' untranslated region. If no N-terminal ID sequence is used, the polymerization may occur immediately after the ATG initiation sequence or preferentially after the ATGG sequence--which preserves both the consensus vertebrate initiation site and the NcoI site. The most commonly used enzyme for this polymerization is terminal transferase (usually from calf thymus), which is routinely used for generating homopolymeric regions for DNA cloning. However, by mixing different deoxynucleotide triphosphates, semi-random heteropolymers of DNA may be synthesized on a DNA primer with a free 3'-OH. Again, the A, T, C, and G base composition may be manipulated to favor certain codons and reduce the frequencies of nonsense codon by controlling the relative concentrations of the four deoxynucleotide triphosphates. In particular, a lower amount of dATP should reduce the frequencies of nonsense codons (TAA, TAG, and TGA). E. coli DNA polymerase I is reported to carry out non-template (de novo) synthesis of DNA and may be used instead of terminal transferase (A. Kornberg, DNA Replication, W.H. Freeman & Co., San Francisco, Calif., 1980). Other enzymes or chemical methods may also polymerize DNA directly onto the expression units. Second-strand synthesis is most easily accomplished by random primer extension, but other methods may provide the same result. Again, the use of nonsense suppressing tRNAs may greatly assist in overcoming the problem of stop codons in this semi-random DNA sequence.
    III. TRANSCRIPTION OF THE NOVEL GENES
    If DNA expression units are used with the semi-random sequences, mRNA may be easily created with RNA polymerase. As discussed above, T7, T3, and SP6 RNA polymerases are commercially available and extremely active. As an example, a DNA expression unit with a T7 promoter is treated with T7 RNA polymerase according to manufacturers' specifications. Approximately 50 mRNA copies may be synthesized routinely for each DNA molecule in 30 minutes. The DNA may be degraded with RNase-free DNase. If the original DNA library had a sequence diversity of 1012 molecules, the resulting mRNA pool should reflect the same level of diversity but now contain 50 or more RNA copies of each different DNA molecule. An RNA library of 6 µg may contain 50 copies of 1012 different mRNAs that are each capable of expressing a semi-random polypeptide of 30 amino acids. Since 6 µg is easily manageable in small test tubes, standard laboratory tools and vessels may be used.
    The 5' ends of mRNAs need to be modified with the addition of diguanosine triphosphate "caps" (or analogs) for efficient translation in eukaryotic systems. The 5' capped mRNA may be generated during in vitro transcription (Hope and Struhl, Cell 43:177-188, 1985) and/or in the in vitro translation process (Krieg and Melton, Nucleic Acids Res. 12:7057-7070, 1984). To cap messages during transcription, an excess of diguanosine triphosphate or an analog thereof (m7G(5')ppp(5')G, from Boehringer Mannheim Biochemicals, for example) is used during the RNA polymerization relative to GTP. An mRNA capping kit based on this method is commercially available from Stratagene (California), which claims that 90%-95% of the resulting RNA is capped.
    If the expression unit is RNA-based, such as the QB replicase system, a few RNA copies may be generated with T7 or other promoter systems (see Lizardi et al., ibid, 1988) if the novel gene constructions involve a DNA plasmid. Once RNA copies exist (or if the novel genes were assembled at the RNA level), RNA-directed RNA polymerase is capable of making a virtually unlimited number of copies of the RNA library (one billion copies are easily attainable). However, the diversity of the library remains the same. With RNA phages, such as QB, the library may be self-sustaining at the RNA level without the necessity of going through a DNA intermediate.
    IV. TRANSLATION OF THE RNA
    Several in vitro translation methods are widely known. For convenience, the rabbit reticulocyte or wheat germ systems may be used with minor modifications. In vitro translation kits are available commercially. For example, the "Translation Kit, Reticulocyte, Type I" from Boehringer Mannheim Biochemicals has all components for 100 translation reactions. Each reaction has been optimized for approximately 1 µg of mRNA in a 25 µl volume. One µg of mRNA is sufficient to code for over 4 x 1012 novel genes, as described above. Therefore, it is possible to translate extremely high numbers of novel genes in relatively small volumes. For example, 1013 80S ribosomes only weigh approximately 66 µg. Because of the small size of the mRNA, only a few ribosomes per message are expected to saturate the mRNAs.
    As described in the protocol for the representative translation kit noted above, GTP and m7G(5')ppp(5')G are required for the efficient translation of in vitro transcribed RNA. Even if mRNA capping has been previously performed during transcription, as described above, it may be advantageous to add the diguanosine triphosphate (or analog thereof) and guanylyltransferase (Krieg and Melton, ibid., 1984) to the translation reaction. In the absence of capping during transcription, the two reagents are necessary for the efficient translation of the mRNA. In particular, when QB constructions are translated, diguanosine triphosphate (or analog thereof) and guanylyltransferase may be necessary for capping the RNA molecules during translation.
    Other techniques may also be employed to optimize translation and especially ribosome attachment to the mRNAs. For instance, it may be desirable to add ribonuclease inhibitors, such as heparin. Eukaryotic systems, such as the wheat germ and reticulocyte translation methods, may yield similar results to prokaryotic systems. The prokaryotic systems have the advantages of smaller ribosomes and more readily available nonsense suppressor tRNAs. In addition, in prokaryotic cells transcription and translation are often simultaneous reactions. In the absence of coupled transcription and translation in prokaryotes, mRNA stability is greatly reduced. Therefore, a prokaryotic in vitro expression system may be used which combines transcription and translation.
    As described above, a preferred embodiment of the present invention is the use of suppressor tRNAs (especially tyrosine-inserting suppressors), which may be produced through recombinant DNA technology and/or by the partial purification of these molecules from mutant cell lines. Radioactive amino acids, especially S35-methionine, may be useful for monitoring in vitro translation and for following low amounts of polysomes in subsequent steps.
    After about 30-60 minutes, protein synthesis begins in the translation reactions. The precise time may be determined for any given set of translation conditions by the use of radioactive amino acids (such as S35-methionine) and monitoring TCA precipitable counts, which is indicative of polypeptide synthesis. After the onset of protein synthesis, cycloheximide at a final concentration of 1 µg/ml is added to prevent the movement of the ribosomes on the mRNAs (Lynch, Meth. Enzym. 152:248-253, 1987). This level of cycloheximide and a Mg+2 concentration of 5 mM may be use to maintain the mRNA-80S ribosome-nascent polypeptide complexes (polysomes). Other ribosome inhibitors may also be used since cycloheximide, for example, will not work on prokaryotic ribosomes. However, in the absence of GTP the polypeptide release from the ribosomes should not normally occur.
    V. BINDING POLYSOMES TO SUBSTANCES OF INTEREST
    The list of potential compounds to which the nascent peptide might bind is virtually unlimited. The coupling chemistries to link these compounds to columns, matrices, filters, beads, etc., will depend to a great degree upon the nature of the compound. In some cases, whole cells or cellular fractions may be used to find peptides which bind to cellular components, such as receptor proteins and other membrane-bound molecules.
    For many proteins and nucleic acids, binding to nitrocellulose or similar artificial surfaces is a property of the filters or fibres. In these cases, the substances of interest are "stuck" to the membranes by established protocols. Bovine serum albumin (BSA), gelatin, casein or nonfat milk, or other proteinaceous material is then typically added in excess to bind up any "free" surface sites. For example, an antibody is first bound to nitrocellulose by placing a solution of the antibody on a nitrocellulose disk in a microtiter dish. After absorbing the antibody to the nitrocellulose, the disk is washed by moving the nitrocellulose disk to fresh microtiter dishes containing saline. After the washes, the disk is placed in a microtiter dish containing gelatin in solution. The disk is then washed again with saline.
    Before allowing the polysomes to bind substances of interest, it may be desirable to pre-absorb the polysome mix against BSA, gelatin, and in particular the proteinaceous material (blocking protein) used in excess as described above. In this manner, polysomes which bind to the blocking protein or nonspecifically to any protein are removed. This pre-absorption step will lead to much greater specificity of polysomes binding to the substance of interest. For binding to specific antibodies (as in the case above), the pre-absorption step(s) may include another antibody, preferably of a similar subclass, but having different variable/hypervariable regions. By screening out polysomes which bind generally to antibodies but not to the variable/hypervariable region, the present invention may be useful for selecting anti-idiotypic binding proteins. Such molecules may have biological or enzymatic activity (as seen for some anti-idiotypic antibodies) or be useful as vaccines.
    The binding of polysomes to substances of interest may be accomplished in the presence of MgCl2 (5 mM) and RNase inhibitors, such as heparin. In addition, specific incubation parameters--such as low or high temperature, high or low salt, or different pHs--may be used to locate polypeptides which bind conditionally, depending on the environment. Incubation times will depend upon the concentration of the bound substance of interest and upon the nature of such substance.
    VI. ISOLATION OF POLYSOMES WHICH BIND TO SUBSTANCE(S) OF INTEREST
    After allowing the polysomes to selectively bind to the substance(s) of interest, nonbinding polysomes are generally removed by washings. This wash should contain MgCl2 and perhaps gelatin, BSA, or other proteins to help reduce nonspecific binding of polysomes. If radiolabeled amino acids are used in the translations, washes (serial or flow-through) should continue until little detectable change is observed in radioactive counts bound to the substance of interest. If the amino acids are not labelled, washes should continue until at least 10-6 dilution of the polysome solution is obtained.
    Conditionally-binding novel peptides may be isolated after these washes by shifting the polysomes into the desired environment for nonbinding, such as higher temperature, different pH, high metal ion concentration, or low salt concentration. Those peptides (and their attached ribosome mRNA complexes) which do not bind under the second ("stringent") condition(s) will be released into the solution and represent potential conditionally-binding factors against the substance of interest. Once immobilized, conditionally-binding peptides may be used to purify substances of interest. Alternatively, conditionally-binding peptides may serve as reagents in monitoring environmental changes.
    VII. DISRUPTION OF THE ISOLATED POLYSOMES
    The isolated (bound) polysomes may be easily disrupted by the removal of Mg+2 (by dilution or via chelating agents) or through the destruction of proteins by a number of methods (proteases, chloroform, etc.). Although dilution is the easiest method, it may not result in as thorough a disruption of the polysomes as compared to other methods. The bound polysomes are placed in a solution lacking Mg+2 to liberate the mRNA; RNase inhibitors may be desirable.
    Conditionally-binding polysomes, which were released under any of the desired environments, may be treated in a similar fashion to disrupt the polysomes and release their mRNAs.
    VIII. RECOVERING MESSENGER RNA AND CONSTRUCTING cDNA
    Theoretically, if a single polysome binding to the substance of interest carries a mRNA, its rare mRNA is capable of being isolated (recovered) from the entire library of mRNAs. The mRNA may also be amplified by several techniques in order to facilitate isolation.
    The use of the polymerase chain reaction (PCR) on a single copy of DNA and on rare mRNA is well documented. (For review, see H.A. Erlich (ed.), PCR Technology, Stockton Press, New York, N.Y., 1989; M.A. Innis et al. (eds.), PCR Protocols: A Guide to Methods and Applications, Academic Press, San Diego, Calif., 1989; H.A. Erlich (ed.), Polymerase Chain Reaction: Current Communications in Molecular Biology, Cold Spring Harbor Press, Cold Spring Harbor, N.Y., 1989.) Briefly, the rare mRNA is first subjected to cDNA synthesis by standard means. Since the sequences of the 5' and 3' regions are known, specific primers may be used for cDNA synthesis. Second, the single cDNA may then be amplified through the use of specified primers (even the same primers as those used in cDNA synthesis). The primers used for PCR may include sequences which restore the 5' and 3' regions of the original expression unit--that is, sequences which restore the promoter (e.g., the T7 polymerase recognition sequence) and 3' region are desirable. By recreating the expression unit in this manner, repeated rounds of transcription-translation-polysome selection may be performed until virtually all of the selected genes code for binding peptides. For expression units based on RNA phages, such as QB, recovery and amplification of the rare mRNA is simplified because each mRNA may be capable of replication to one billion copies or more, using the appropriate replicases.
    IX. EXPRESSION OF NOVEL GENES
    Once the novel genes have been isolated and sequenced, they or related sequences may be (1) cloned, (2) chemically reproduced, (3) mutated, and (4) expressed by protocols well known in the art. Large-scale production of the novel polypeptide may be accomplished through recombinant DNA methods, using genetically engineered microorganisms. A large variety of prokaryotic and eukaryotic expression systems exist for the in vivo synthesis of the novel binding peptide. The convenient NcoI site described above or other restriction sites may be used to connect the coding region of the novel gene to a desired promoter. It will be evident to those skilled in the art that other gene splicing strategies exist as well. A translation stop codon and a transcription termination sequence may be added to the 3' end of the novel gene for proper expression of the gene in a microorganism. This genetically engineered sequence may then be placed on a plasmid or vector and placed within a desired host cell by transformation, transduction, infection, electroporation, microinjection, or other similar methods. The novel peptide sequence may be attached to a signal sequence for possible secretion from the microorganism and/or may contain ID peptides, as herein described, for quantifying and purifying the resulting gene product. The novel peptide or related sequence may be attached to other translated sequences to form a hybrid or fusion protein which is similarly expressed in a genetically engineered organism. Alternatively, large-scale in vitro transcription and translation methods may be used to produce commercial quantities of the polypeptide.
    Finally, if the amino acid sequence of the novel peptide is relatively short, currently available technologies allow for the large-scale chemical synthesis of the polypeptide. Chemical synthesis of the novel peptide has advantages over the in vitro and in vivo expression systems. Among these advantages chemical synthesis (1) is a better defined and therefore more reproducible system for synthesis, (2) has no contaminating sources of DNA and RNA, (3) has no contaminating sources of nucleases, proteases, and other modifying enzymes, and (4) provides a relatively pure product after synthesis.
    X. REPETITIVE ENRICHMENT FOR SPECIFIC POLYSOMES
    Depending upon the amount of background, nonspecific binding of polysomes to the substance(s) of interest, one may choose to perform a few to many cycles of translation-transcription-binding-recovery as described above to increase the frequency of sequences which code for the desired polypeptide. For example, if each cycle increases the frequency of the desired novel gene(s) by 104, then three cycles may be sufficient for isolating a sequence which exists in the original library at a frequency of 10-12. Each cycle may be completed in one to three days; and many steps of the process may be performed by automated workstations, or robots. Therefore, many cycles may be routinely accomplished for a desired binding activity within one week.
    XI. SCREENING FOR ACTIVITIES OF TRANSLATED PRODUCTS WITHOUT POLYSOME BINDING
    One aspect of the present invention does not require polysome binding for gene isolation. Instead, in vitro translation is allowed to proceed to completion, with the resultant release of the new polypeptides from the ribosome. This is accomplished by the use of nonsense codons or by the ribosomes "falling off" the end of the mRNAs. The new peptides may be separated from the ribosomes and other components of the translation reaction by gel filtration and/or centrifugation and/or other means, in order to concentrate the translation products. The peptide mixture is then challenged to exhibit biological or enzymatic activity--for example, the peptides are assayed for mitogenic activity by treating tissue culture cells lacking a growth factor.
    If biological or enzymatic activity is observed within the entire array or subset of the novel peptides, the gene(s) which codes for this activity may be located by subdividing the original library or an RNA copy of the library and screening for activity in a subdivision. After successive subdivisions, the desired gene may be isolated to a pool containing (for example) less than 1,000 different sequences. In theory, the desired gene may be completely isolated by subdivision (to a "pool" containing just that one gene). With PCR, QB replicase or other methods (as described above), the desired sequences may be amplified to a level where in vitro transcription and translation produces a highly enriched peptide solution having the biological/enzymatic activity. At a frequency of 1 to 10-3, the gene of interest may be readily isolated and cloned into appropriate expression systems, using methods currently available.
    XII. CELL-FREE IDENTIFICATION OF NOVEL GENES AND PEPTIDES
    After a novel gene with putative binding or biological activity has been isolated, it may be demonstrated that the purified sequence codes for the activity of interest by amplifying the DNA and/or RNA so that sufficient mRNA is produced for larger-scale in vitro translation. The translation products of this purified sequence should be nearly homogeneous polypeptides having the assayable activity. The gene and/or the polypeptide may be sequenced by existing methods to establish the composition of the novel polypeptide. Alternatively, the purified gene may be cloned into microorganisms for amplification and expression. Subsequently, biological/binding activities as well as sequence identity may be established for the novel gene and polypeptide.
    XIII. CREATING NOVEL HYBRID PROTEINS
    After the nucleic acid sequence has been determined for the novel gene, this sequence may be incorporated into larger genes to create hybrid proteins, which have the characteristics of the novel peptide and other desirable properties. One class of hybrid proteins which may be created by this technology is characterized by specific binding to cells and cytotoxic abilities. For example, a cell surface receptor-binding peptide may be joined to ricin or other toxins via DNA splicing methods. This type of hybrid protein may be used to selectively kill different cell types, including pathogens and tumor cells. The gene which encodes the hybrid protein may be completely synthesized or result from splicing the appropriate gene fragments together. This gene may be expressed in a variety of expression systems.
    A preferred embodiment of this invention is the replacement of variable and hypervariable regions of antibody and antibody-like genes by novel gene sequences which code for binding activities against substances of interest. In this manner, a much greater range of diversity is possible against antigens of interest; and the screening process may be much more efficient and time-saving than the production methods for monoclonal antibodies against the same antigens. These "custom" hybrid antibody genes may be expressed in a number of organisms to produce active antibodies with new specificities or properties.
    XIV. OTHER COMMERCIAL USES OF THE INVENTION
    The application of the present invention in diagnostic tests parallels the use of monoclonal/polyclonal antibodies, and is more advantageous, primarily because the isolation of novel polypeptides as herein described may take considerably less time (one week versus a few months for antibodies). In addition, other advantages may be seen. The novel polypeptides may be considerably smaller molecules than the antibodies. Therefore, synthesis, purification, and/or manufacturing of the novel peptides may be greatly simplified and cost-effective as compared to antibodies. The smaller size may also aid in stability, formulation, and in reaching the target molecules.
    The novel polypeptides may be identifiable by (1) fusing them to a biologically active peptide which has a quantifiable activity (such as peroxidase or other enzymatic activity), (2) synthesizing them with an ID peptide, described above, to which existing antibodies are known to bind, (3) radioactively labelling them, (4) chemically adding markers, such as fluorescent dyes or metallic substances, or (5) any combination of the above. To increase specificity in the diagnostic use of the novel polypeptides, two or more different polypeptides may be used. In addition, novel polypeptides may be used as competitive binding elements in diagnostic tests which rely upon competitive binding to antigens or substrates.
    Another advantage of novel polypeptides generated via the present invention is that they may bind to many classes of molecules which would not elicit a strong immune response, because some molecules are not complex enough or are too similar to an organism's resident compounds to trigger antibody formation. In addition, the use of novel polypeptides in diagnostic binding assays may have a much greater scope than the traditional antibody-based methods.
    The novel polypeptides of the present invention may also be used therapeutically as originally isolated or as part of fusion proteins. For example, if a novel polypeptide were selected to bind a given toxin, it might also neutralize the toxin. If a new polypeptide is bound to a viral receptor site on a cell membrane or to the virus's attachment mechanism, infection of the cell may be diminished. As described earlier, fusion proteins carrying novel polypeptide recognition sequences in addition to a toxin may be used to selectively kill diseased or malignant cells. The binding of novel sequences to infected or malignant cells may trigger an immune response against the cell-peptide complex and, therefore, may be useful in the control of disease.
    EXAMPLES
    The following examples are provided by way of illustration and not by way of limitation. Within the examples, standard reagents and buffers that are free from contaminating activities (whenever practical) are used. It is preferred to exercise care to avoid ribonucleases and PCR product contamination.
    EXAMPLE 1 SYNTHESIS OF A NOVEL GENE LIBRARY
    The sequences and strategies for creating a novel gene library require careful planning by those skilled in the art. The 5' untranslated region of the expression unit contains an RNA polymerase site, a ribosome binding site, an initiation codon, and selected 5' untranslated sequences. The polymerase binding site used in this example is the T7 promoter sequence: TAATACGACTCACTATAGGGAGA (23-mer), which is placed at the 5' end of the expression unit.
    A rabbit reticulocyte system is used for translation of the RNAs synthesized from the T7 promoter. Therefore, the ribosome binding site should include at least part of the consensus sequence for eukaryotic untranslated regions. In her review article, Kozak (ibid., 1987) suggests that very short untranslated regions (less than 10 nucleotides) do not initiate protein synthesis efficiently. A selected untranslated region of 36 nucleotides is used here. This untranslated region is derived from the naturally-occurring (36-base pair) upstream sequence of the adult rabbit hemoglobin (alpha-globin):
    ACACTTCTGGTCCAGTCCGACTGAGAAGGAACCACCATGG, where the underlined ATG represents the start of translation at a methionine initiation codon (Baralle, Nature 267:279-281, 1977). The rabbit alpha-globin untranslated sequence is chosen because (1) it is expected to be a favorable substrate in a rabbit reticulocyte system and (2) it contains the important "motifs" of Kozak's model mRNA.
    The alpha-globin sequence is modified in the following ways for in vitro gene expression. First, the 5' A (underlined above) is replaced by a G, which may aid in the capping of the mRNAs (Green et al., Cell 32:681-694, 1983). Second, the G (underlined in the alpha-globin sequence) is replaced with an A to help eliminate a putative secondary structure in the untranslated region of alpha-globin which is hypothesized to reduce the initiation of protein synthesis by 60% relative to the beta-globin mRNA (Baralle, ibid., 1977). This second change also creates a convenient GATC restriction site in the 5' untranslated region. The resulting leader sequence, including the ATGG of the coding region, is therefore the following:
    Figure 00300001
    This leader sequence is placed immediately downstream from the T7 promoter.
    The 3' region contains (1) a selected sequence for specific-primer-directed DNA synthesis, (2) a GGG-rich region which codes for a polyglycine tether that gives the nascent polypeptide spatial freedom to bind the substance of interest, and (3) convenient restriction sites whose resulting RNA secondary structure may impede the translocation of ribosomes off the mRNA. The polyglycine region comprises 20 codons for glycine; most of the glycine codons are adjacent GGG triplets, which code for glycines in all reading frames. However, some of the glycine codons are GGT or GGA to keep the DNA strands in proper register. The restriction sites for Bam HI (GGATCC) and NotI (GCGGCCGC) are chosen to be placed very near the 3' end of the gene; in the mRNA these sequences are expected to form hairpin loops. To prevent second-strand self-priming (of hairpin loops) by the NotI sequence, an addition of AAAA is made at the 3' end. The 3' region therefore has a general sequence of (GGG or GGT/A)20 followed by GGATCCGCGGCCGCAAAA. A specific sequence for this region is given below.
    The semi-random gene sequence is synthesized with known 5' and 3' ends which undergo basepairing and ligation with the fully described 5' untranslated region and 3' region segments. To achieve this end, the semi-random gene is synthesized with a 5' CACCATGG, which may basepair with the octamer CCATGGTG on the complementary strand of the 5' untranslated region. The initiation (first) codon, ATG, is necessary for translation of the semi-random sequences. The subsequent G is the first position of the second codon and is constant to preserve the NcoI site at the front end of the gene. The rest of this second codon and the next 28 codons are synthesized following the rules outlined earlier for reducing nonsense triplets. That is, in the first codon position, equal molar amounts of C, A, and G are used but only half that amount of T is used. In the second codon position, the amount of A is reduced to half of the level of the other three bases. In the third codon position, only G and C are used, and in equal molar amounts.
    After codon 30 is synthesized, GGTGGGGG is added. This sequence codes for two glycine residues and is used to ligate the semi-random sequences to the 3' region, which has a complementary CCCCCACC overhang on the opposite strand. The result of this synthesis is a sequence that codes for virtually all 30 amino acid polypeptides (beginning with methionine) and has a polyglycine tether. The probability of no stop codons in this string of triplets is approximately 80%. By using partially purified yeast tyrosine-inserting UAG suppressor tRNA (Pelham, ibid., 1978) during the subsequent translation, over 90% of the semi-random sequences are expected to code for full-length polypeptide.
    The specific oligonucleotides to synthesize are listed below:
  • I. T7 Promoter & "Globin" Leader (for gene synthesis and PCR):
    Figure 00320001
  • II. Anti-T7 Promoter & "Globin" Leader (for gene synthesis):
    Figure 00320002
  • III. Semi-Random Gene (for gene synthesis):
    Figure 00320003
  • IV. Poly-Glycine & 3' Restriction Sites (for gene synthesis):
    Figure 00320004
  • V. Anti-Poly-Glycine & 3' Sites (for gene synthesis):
    Figure 00320005
  • VI. Anti-Poly-Glycine & 3' Sites (for cDNA synthesis and PCR):
    Figure 00320006
  • Sequences I and II are mixed in equimolar amounts in standard TE Buffer and heated at 65°C for 5-10 min. The complementary sequences (which comprise the 5' untranslated region) are allowed to anneal at 50°-60°C for one hour or longer, are allowed to cool slowly to room temperature, and are thereafter stored at 0°-4°C. Sequences IV and V are likewise treated to form the double-stranded 3' region. These duplexes each have an eight-base, single-stranded overhanging sequence which is complementary to the known ends of Sequence III.
    Equimolar amounts of I/II duplex, IV/V duplex, and semi-random Sequence III are ligated with T4 DNA ligase overnight at 13°-15°C in Ligase Buffer. The ligation mix is then run on a 1.5% agarose gel to separate out the desired ligation product, which is approximately 200 base pairs (233 bp if completely double-stranded, which it is not). The "200 bp" DNA band is gel purified with NA45 paper (S&S) or by any of several protocols. A total of 2.5 µg (representing approximately 1013 DNA molecules) or more is desirable.
    Complete double-stranded synthesis of novel genes is accomplished with DNA Polymerase I, Klenow, using standard methods. The double-stranded 3' region provides a primer for the "second-strand" synthesis of the semi-random sequences. T4 DNA ligase is used to join the newly synthesized DNA to Sequence II, thereby filling the nick in the second strand. The DNA library is phenol/chloroform extracted and ethanol precipitated.
    10 µg of completely double-stranded DNA molecules has a sequence diversity of 4 x 1013. This library may then be transcribed with T7 RNA Polymerase to yield translatable mRNAs. However, with each transcription, the DNA library is consumed, unless DNA copies are made. To replicate the DNA library, 100 ng aliquots are each distributed to 500-µl tubes for PCR amplification in 200-µl reactions. According to PCR Technology, pp. 18-19 (Erlich, ibid., 1989), each 200-µl PCR reaction yields about 5.2 µg of DNA--or an approximately 50-fold duplication of DNA in each aliquot. The aliquots are pooled. The pooled sample contains on the average 50 copies of each semi-random sequence and therefore may be used repeatedly (50 times, for example) without a large loss of diversity for each translation with T7 RNA Polymerase. If the library is to be replicated with PCR, then the Klenow filling and ligation steps, described above, may be unnecessary, since the Taq polymerase is capable of filling in the gap and nick-translating DNA (D. H. Gelfand, PCR Workshop, Society of Industrial Microbiology Meeting, Seattle, Wash., 1989). After nick translation, the gene is double-stranded and able to be PCR amplified.
    Examples of oligonucleotide primers for PCR amplification of the DNA library are listed above in sequences I and VI. Generally, oligonucleotides of 25-30 bases are used for PCR amplification; however, longer primers may be used. It is important that the primers do not share significant homologies or complementary 3' ends. Sequences I and VI have noncomplementary ends and no obvious regions of extensive homology.
    In addition, after translation of these novel gene sequences, the resulting mRNAs lack T7 promoter sequences. Sequence VI is used as the primer for first-strand cDNA synthesis . Sequence I is used as the primer for second-strand synthesis and restores the T7 promoter to the cDNA. In this way, later rounds of translation are possible on the selected novel gene sequences. PCR amplification may be necessary if the resulting cDNAs are relatively rare.
    EXAMPLE 2 TRANSCRIPTION OF NOVEL GENES
    The DNA library (or a representative aliquot of those sequences) described in Example One is transcribed with T7 RNA polymerase. 2.5 µg of this DNA codes for nearly 1013 different polypeptides. The DNA is capped during transcription with Stratagene's mCAP™ Kit, according to the manufacturer's specifications. Approximately 5-10 µg of mRNA is expected. Generally, with T7 RNA polymerase, nearly 10 times this level of RNA is synthesized; however, the conditions for the capping reaction limit mRNA production in this case. The DNA is removed with DNase I, provided in the kit. The capped mRNA is phenol/chloroform extracted and precipitated with ethanol. The RNA is resuspended in 10 µl of TE and stored at 0°-4°C.
    EXAMPLE 3 TRANSLATION OF NOVEL GENES
    The capped mRNA is translated with Boehringer Mannheim Biochemical's rabbit reticulocyte kit, with all 20 amino acids at 312.5 µmol/l each. Capped mRNA from Example 2 is added to each reaction at 0.5 µg per reaction and is treated according to the manufacturer's protocol. After around 60 minutes at 30°C, cycloheximide is added to a final concentration of 1 µg/ml. MgCl2 is adjusted to 5 mM, and heparin is added to 0.2 mg/ml. The reactions are pooled and submitted to a discontinuous sucrose gradient, according to Lynch (ibid., 1987). The polysomes may be frozen at -70°C or used directly.
    EXAMPLE 4 IMMOBILIZATION OF ANTIBODIES AS THE SUBSTANCE OF INTEREST
    Antibodies may be used to select for novel binding peptides. Peptides which bind to the hypervariable/variable regions of the antibodies ("anti-id peptides") may behave like the original epitopes which were used as immunogens. Because the novel anti-id peptides may mimic the original epitopes, these peptides may be useful as vaccines and/or may demonstrate biological activities, in much the same way that anti-id antibodies have been shown to have biological (sometimes catalytic) activities.
    Examples of useful antibodies are anti-fibronectin, anti-nerve growth factor, anti-CD4, and anti-tumor necrosis factor, which are all available from Boehringer Mannheim Biochemicals. In general, antibodies to receptor molecules, growth factors, surface antigens, and biologically active peptides, as well as neutralizing antibodies to toxins and diseases, are good candidates for which to isolate anti-id binding peptides that may have agonist or antagonist properties or serve as vaccines.
    The antibodies are affixed to Immobilon" PVDF (polyvinylidene difluoride) membrane from Millipore Corporation, according to Pluskal et al. (BioTechniques 4 :272-283, 1986). For example, anti-fibronectin antibody (from clone 3E3, Boehringer Mannheim Biochemicals) is absorbed onto a 0.5 cm x 0.5 cm square of PVDF, that has been "wetted" with 100% methanol and washed twice with 0.9% (w/v) NaCl in 10 mM Tris buffer pH 7.4 (Saline Buffer). The amount of antibody needed is dependent upon the binding parameters of the desired anti-id peptides(s); Immobilon" PVDF is reported to bind 172 µg/cm2 of IgG. For convenience, 1 µg of anti-fibronectin IgG1 in saline buffer is absorbed onto the PVDF square by incubating at room temperature for at least two hours. The PVDF is then washed with the Saline Buffer twice. The membrane is next incubated with a "blocking solution," containing 5% (w/v) gelatin in saline buffer for at least two hours at room temperature, so that the gelatin is absorbed into unoccupied sites of the PVDF. The membrane is then washed twice with 0.1% gelatin in saline buffer. A similar treatment is done with 10 µg anti-keratin antibody (from clone AE1, Boehringer Mannheim Biochemicals), which is the control IgG1 as described below.
    EXAMPLE 5 POLYSOME BINDING TO ANTIBODIES
    Polysomes with nascent semi-random peptides are incubated in 1-ml reactions, each containing PS Buffer (0.9% NaCl, 10 mM Tris pH 7.4, 1% gelatin, 15 mM MgCl2, 0.2 mg/ml heparin, and 1 µg/ml cycloheximide) and a PVDF square with 10 µg anti-keratin IgG1, described in Example 4. This pre-absorption step is done at 0°-4°C with gentle agitation for four hours to select out nonspecific binding of polysomes to gelatin and IgG1. The anti-keratin PVDF square is removed with jewelers' forceps and is replaced with the anti-fibronectin PVDF square. The mixture is incubated for four more hours under the same conditions to allow specific polysome binding to the variable/hypervariable region of the anti-fibronectin antibody. The anti-fibronectin PVDF square is removed and washed three times by transferring it serially to fresh PS buffer.
    EXAMPLE 6 RECOVERING NOVEL GENES WHICH CODE FOR ANTI-ID PEPTIDES FROM POLYSOMES
    The PVDF membrane, which holds the washed antibody-bound polysomes, is transferred to a tube containing 100 µl of 0.1 mM EDTA and is gently shaken at room temperature for 5-10 minutes to disrupt the polysomes and liberate mRNA. The PVDF is removed, placed in a fresh tube of 0.1 mM EDTA, and stored at 0°-4°C overnight or longer (as a back-up). The released mRNA from the first EDTA treatment is reverse transcribed; and the resulting cDNA is amplified, according to PCR Technology (ibid., 1989), p. 91, with slight modification. Instead of using random hexamer for priming the cDNA synthesis, a sequence complementary with the known 3' region (such as Sequence VI listed earlier as the downstream primer) is used for both cDNA synthesis and PCR reactions. The reverse transcriptase step is done in 100 µl of PCR buffer with the appropriate relative amounts of the other reagents (instead of 20-µl reaction). After the reverse transcriptase reaction, the mixture is split into 20 µl aliquots; and each aliquot is amplified as described in PCR Technology, using Sequence I or a similar DNA upstream primer. After PCR amplification, the five aliquots are pooled, phenol/chloroform extracted, and ethanol precipitated. This cDNA is then resuspended in TE and stored at 0°-4°C.
    The selected DNA is transcribed with T7 RNA polymerase and translated in a reticulocyte system, as previously described. In this case, the desired sequences are greatly amplified compared to the original DNA library. By repetition of this cycle, which is greatly aided through the use of programmable workstations, desirable novel genes are concentrated to a level where conventional cloning and expression methods are practical. In addition, by dilution to low Poisson Distribution of genes, a single novel gene(s) may be isolated, amplified, transcribed, and translated to demonstrate specific binding capability of the gene product(s). Once binding has been demonstrated, the isolated gene(s) and polypeptide(s) may be sequenced for identification.
    After the sequence of the novel binding peptide is known, many methods exist for the manipulation and large-scale synthesis of the peptide, as described herein.
    EXAMPLE 7 COMPETITION ASSAY FOR BINDING PEPTIDES
    After novel genes which code for binding peptides are selected, the amplified pools of recovered cDNA are assayed for the presence of the genes. Where ID sequences have been intentionally included to be coexpressed with the semi-random DNA sequences, ELISAs or other immunological assays for the known part of the peptide are used to detect the binding of the novel portion of the peptide to the substance of interest. However, when no ID sequence is present and/or a confirmation of binding specificity is desirable, competition assays for the peptides are carried out. Competition assays, including competition ELISA tests, are used to monitor for the presence of binding sequences within the various cDNA pools generated by the present invention.
    One example is the screening of the cDNA pools for genes which encode peptides which bind anti-Pseudomonas exotoxin (anti-PE) antibody. After two rounds of selection for polysome binding to the anti-PE antibody, different aliquots of the resultant cDNA pool were each transcribed in a 200-µl reaction with T7 RNA polymerase (30 units) under standard conditions, starting with approximately 200 ng of DNA. The mRNA products were phenol/chloroform/isoamyl alcohol extracted and precipitated with sodium acetate and ethanol at -20°. The precipitates were each centrifuged and resuspended in 16 µl of distilled water which had been treated with diethylpyrocarbonate to remove nucleases.
    The resuspended mRNAs were heated to 65° for five minutes and then placed on ice. The RNAs were translated with a wheat germ kit (Boerhinger Mannheim Biochemicals) according to the manufacturer's recommendations. Each RNA sample was expressed in a 50 µl translation reaction with 25 microcuries of 35S-methionine and 0.5 µl of RNase inhibitor. The reactions were run for 15-60 minutes at 30°. At the end of the translation, the samples were each equally divided: one half was used to bind the substance of interest without competing substrate, while the other half was used to bind the substance of interest in the presence of excess competing substrate. In this case, the substance of interest was anti-PE antibody. The competing substrate was a 14-amino acid peptide (PE peptide), which is derived from the toxin protein sequence and known to bind the antibody. The PE peptide sequence is Val-Glu-Arg-Leu-Leu-Gln-Ala-His-Arg-Gln-Leu-Glu-Glu-Arg. See Wozniak, et al., Proc. Natl. Acad. Sci., 85: 8880-8884 (1988).
    The competition assays were done over ice in 96-well microtiter dishes with flat bottoms. Immobilon PVDF disks were made with a standard 0.635 cm (1/4 inch) holepunch and placed in wells labelled "A". 50 µl of methanol were added to the disks in "A" to wet and sterilize the membranes. The disks were transferred with forceps to wells "B" which contained 200 µl of Saline Buffer plus 10 mM MgCl2 (TSM buffer). The disks were further washed by moving them to wells "C" which also contained 200 µl of TSM. They were then transferred to wells "D" which contained 25 µl TSM plus 3 µl of anti-PE antibody (4.6 µg/µl). The antibody was absorbed to the disks for three hours on ice with gentle rotation (50-100 RPM on a platform shaker). Afterwards, 75 µl of 2% nuclease-free BSA was added to "D" and absorbed for 1 hr. at 100 RPM.
    The disks were washed twice in 200 µl of TSM plus 0.1% BSA (in wells "E" and "F") for 30 minutes in each well and were then ready for peptide binding. In wells "G" 26 µl of TSM plus 0.1% BSA was mixed with 25 µl of each translation reaction described above--half of the 50 µl wheat germ system. Into one-half of each of the "G" wells, 1 µl of PE peptide (1 mg/ml in TSM) was added to competitively inhibit the binding of novel radioactively-labelled peptides to the antibody; these wells were labelled "+ Peptide." Into the control "G" wells, 1 µl of TSM was added and the wells labeled: "No Peptide." The disks were added to the appropriate "G" wells and incubated to three hours at 100 RPM on ice for peptide binding to the immobilized antibody.
    After the binding reaction each disk was serially washed eight times in 200 µl of fresh TSM at 0°, with a 10 minute incubation for each wash. The bound radioactivity for each disk was measured in a liquid scintillation counter with a 1 ml cocktail of Ecoscint. The following table lists the results of competition assays on different aliquots of cDNAs obtained from the binding of polysomes to the anti-PE antibody:
    SAMPLE CPM 35S-MET
    WE1 + Peptide 6969
    WE1, No Peptide 8337
    WE2 + Peptide 6163
    WE2, No Peptide 7693
    WP1 + Peptide 5792
    WP1, No Peptide 6303
    WP2 + Peptide 5845
    WP2, No Peptide 6398
    In each case the competing PE peptide reduced the amount of binding of the radioactively-labelled translation products of the selected cDNA pools, compared to the No Peptide controls. These results indicate the presence of gene sequences which code for binding peptides to the anti-PE antibody. Isolation and characterization of these DNA sequences is then done by cloning individual genes into plasmids, such as pUC18, pUC19, Bluescript, and many other available vectors.
    The invention is not to be limited except as by the following claims.

    Claims (22)

    1. A method for isolating a nucleotide sequence which encodes a polypeptide of interest, comprising
      transcribing or replicating an in vitro expression unit which comprises a 5' untranslated region containing a RNA polymerase binding sequence, a ribosome binding sequence, a translation initiation signal, and one or more semi-random nucleotide sequences to produce a mRNA library;
      translating the mRNA library under conditions which maintain polysomes having polypeptide chains attached thereto;
      contacting the polysomes to a substance of interest that is capable of specifically reacting with the polypeptide of interest and isolating mRNA from polysomes that specifically react with the substance of interest.
    2. The method of claim 1, further comprising constructing cDNA from mRNA isolated from the polysomes.
    3. The method of claim 2, further comprising amplifying the cDNA.
    4. The method of claim 2, further comprising expressing the amplified cDNA to produce a polypeptide.
    5. The method of claim 1 wherein said semi-random nucleotide sequence comprises deoxyribonucleic acid or ribonucleic acid.
    6. The method of claim 1 wherein said expression unit includes at least one RNA-directed RNA polymerase recognition sequence.
    7. The method of claim 1, further comprising amplifying the mRNA isolated from the polysomes with an RNA-dependent RNA polymerase.
    8. The method of claim 1 wherein the polysomes that do not bind to said substance of interest are removed by serial dilution or flow-through wash steps.
    9. The method of claim 8 wherein, subsequent to the step of removing non-binding polysomes, the polysomes binding to the substance of interest are exposed to selected stringency conditions such that said polysomes are released from said substance of interest.
    10. A method for producing novel polypeptides by expressing the nucleotide sequence isolated in the method of claim 1, comprising, subsequent to the contacting step of claim 1,
      isolating said polysomes that bind to said substance of interest;
      disrupting said isolated polysomes to release mRNA;
      recovering said mRNA;
      constructing cDNA from said recovered mRNA; and
      expressing said cDNA to produce novel polypeptides.
    11. A method for producing novel polypeptides by expressing the nucleotide sequence isolated in the method of claim 1, wherein, during said translating step, the conditions optionally permit polypeptide chains to be released from the polysomes, and further comprising isolating the mRNA encoding the polypeptides identified in the contacting step by subdividing,
      repeating the steps of transcribing, translating, contacting and isolating by subdividing, such that the gene of interest is isolated ;
      constructing cDNA from said isolated gene; and
      expressing said cDNA to produce novel polypeptides.
    12. The method of claim 11 wherein, subsequent to the step ot subdividing the RNA, amplifying the novel polynucleotides encoding the biologically active polypeptides with polymerase chain reaction or with an RNA-directed RNA polymerase.
    13. The method of claim 1, 10 or 11 wherein the expression unit further comprises a polynucleotide which codes for a selected amino-terminal identification (ID) peptide, said polynucleotide positioned at the 3' end of the initiation codon.
    14. The method of claim 1, 10, or 11 wherein said expression unit further comprises a 3' region of a selected polynucleotide selected from a restriction site, palindromic sequence, sequence encoding an identification peptide, sequence for specific-primer-directed DNA synthesis, a polyA or other homopolymeric polynucleotide sequence.
    15. The method of claim 1, 10 or 11 wherein said expression unit further comprises restriction sites adapted to allow expression of the novel gene in vivo.
    16. The method of claim 1, 10 or 11 wherein said expression unit includes the promoter for T7, T3, or SP6 polymerase in the 5' untranslated region.
    17. The method of claim 1, 10 or 11 wherein the semi-random polynucleotides are generated by mechanically, chemically, or enzymatically fragmenting naturally-occurring DNA or cDNA.
    18. The method of claim 17 wherein the step of chemically synthesizing said nucleotides comprises the steps of (1) utilizing substantially equal molar amounts of C, A, and G, and only half of said substantially equal molar amount of T in the first codon positions; (2) utilizing substantially equal molar amounts of C, T, and G, and only halt of said substantially equal molar amount of A in the second codon positions; and (3) utilizing substantially equal molar amounts of C and G or T and G in the third codon positions.
    19. The method of claim 1, 10 or 11 wherein the step of translating is conducted in the presence of nonsense-suppressing tRNAs.
    20. The method of claim 1, 10 or 11 wherein said substance of interest is selected from a group consisting of surface antigens, receptor proteins, toxins, organic polymers, metabolites, active sites of protein molecules, hormones, antibodies, and the variable/hypervariable region of an antibody.
    21. The method of claim 4, 10 or 11 wherein the step of expressing cDNA comprises in vitro transcription and/or translation of the nucleotide sequence, or cloning the polynucleotide into an expression vector for synthesis in genetically engineered microorganisms.
    22. The method of claim 3 further comprising the step of determining the nucleic acid sequence of the amplified cDNA.
    EP90915566A 1989-10-05 1990-10-04 Cell-free synthesis and isolation of novel genes and polypeptides Expired - Lifetime EP0494955B1 (en)

    Applications Claiming Priority (3)

    Application Number Priority Date Filing Date Title
    US41735789A 1989-10-05 1989-10-05
    PCT/US1990/005682 WO1991005058A1 (en) 1989-10-05 1990-10-04 Cell-free synthesis and isolation of novel genes and polypeptides
    US417357 1995-04-05

    Publications (3)

    Publication Number Publication Date
    EP0494955A1 EP0494955A1 (en) 1992-07-22
    EP0494955A4 EP0494955A4 (en) 1992-08-12
    EP0494955B1 true EP0494955B1 (en) 1998-07-15

    Family

    ID=23653670

    Family Applications (1)

    Application Number Title Priority Date Filing Date
    EP90915566A Expired - Lifetime EP0494955B1 (en) 1989-10-05 1990-10-04 Cell-free synthesis and isolation of novel genes and polypeptides

    Country Status (11)

    Country Link
    US (2) US5658754A (en)
    EP (1) EP0494955B1 (en)
    JP (2) JP3127158B2 (en)
    KR (3) KR0185192B1 (en)
    AT (1) ATE168416T1 (en)
    AU (1) AU638762B2 (en)
    CA (1) CA2067194C (en)
    DE (1) DE69032483T2 (en)
    DK (1) DK0494955T3 (en)
    ES (1) ES2118066T3 (en)
    WO (1) WO1991005058A1 (en)

    Families Citing this family (344)

    * Cited by examiner, † Cited by third party
    Publication number Priority date Publication date Assignee Title
    US5498538A (en) * 1990-02-15 1996-03-12 The University Of North Carolina At Chapel Hill Totally synthetic affinity reagents
    US5747334A (en) * 1990-02-15 1998-05-05 The University Of North Carolina At Chapel Hill Random peptide library
    US5962219A (en) * 1990-06-11 1999-10-05 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: chemi-selex
    US5843701A (en) * 1990-08-02 1998-12-01 Nexstar Pharmaceticals, Inc. Systematic polypeptide evolution by reverse translation
    DE4112440C1 (en) * 1991-04-16 1992-10-22 Diagen Institut Fuer Molekularbiologische Diagnostik Gmbh, 4000 Duesseldorf, De
    US5795747A (en) * 1991-04-16 1998-08-18 Evotec Biosystems Gmbh Process for manufacturing new biopolymers
    AU2313392A (en) * 1991-08-01 1993-03-02 University Research Corporation Systematic polypeptide evolution by reverse translation
    US5783384A (en) * 1992-01-13 1998-07-21 President And Fellows Of Harvard College Selection of binding-molecules
    US5643873A (en) * 1992-05-06 1997-07-01 Affymax Technologies N.V. Peptides and compounds that bind selectins including endothelial leukocyte adhesion molecule 1
    US5728802A (en) * 1992-05-06 1998-03-17 Affymax Technologies N.V. Peptides and compounds that bind selectins including endothelium leukocyte adhesion molecule 1 (ELAM-1)
    US5648458A (en) * 1992-05-06 1997-07-15 Affymax Technologies N.V. Peptides and compounds that bind to ELAM-1
    WO1994005812A1 (en) * 1992-09-02 1994-03-17 The Scripps Research Institute Coupled isothermal polynucleotide amplification and translation system
    US5922545A (en) * 1993-10-29 1999-07-13 Affymax Technologies N.V. In vitro peptide and antibody display libraries
    AU8124694A (en) * 1993-10-29 1995-05-22 Affymax Technologies N.V. In vitro peptide and antibody display libraries
    US5525734A (en) * 1994-06-22 1996-06-11 Affymax Technologies N.V. Methods for synthesizing diverse collections of pyrrolidine compounds
    US5525735A (en) * 1994-06-22 1996-06-11 Affymax Technologies Nv Methods for synthesizing diverse collections of pyrrolidine compounds
    CA2200684A1 (en) * 1994-09-21 1996-03-28 Cytogen Corporation Antigen binding peptides (abtides) from peptide libraries
    US5885577A (en) * 1994-09-21 1999-03-23 Cytogen Corporation Antigen binding peptides (abtides) from peptide libraries
    ATE235504T1 (en) * 1994-12-30 2003-04-15 Chiron Corp METHOD FOR THE CONTROLLED SYNTHESIS OF ANY PEPTIDE MIXTURES ENCODING POLYNUCLEOTIDE MIXTURES
    US6341256B1 (en) 1995-03-31 2002-01-22 Curagen Corporation Consensus configurational bias Monte Carlo method and system for pharmacophore structure determination
    US20010053523A1 (en) * 1995-06-02 2001-12-20 M&E Biotech A/S. Method for identification of biologically active peptides and nucleic acids
    US6537776B1 (en) 1999-06-14 2003-03-25 Diversa Corporation Synthetic ligation reassembly in directed evolution
    US7888466B2 (en) 1996-01-11 2011-02-15 Human Genome Sciences, Inc. Human G-protein chemokine receptor HSATU68
    WO1997045538A1 (en) * 1996-05-31 1997-12-04 Medigene Ag Novel synthetic protein structural templates for the generation, screening and evolution of functional molecular surfaces
    US6875620B1 (en) * 1996-10-31 2005-04-05 Agilent Technologies, Inc. Tiling process for constructing a chemical array
    US8207093B2 (en) 1997-01-21 2012-06-26 The General Hospital Corporation Selection of proteins using RNA-protein fusions
    WO1998031700A1 (en) 1997-01-21 1998-07-23 The General Hospital Corporation Selection of proteins using rna-protein fusions
    US6261804B1 (en) * 1997-01-21 2001-07-17 The General Hospital Corporation Selection of proteins using RNA-protein fusions
    GB2338237B (en) * 1997-02-18 2001-02-28 Actinova Ltd In vitro peptide or protein expression library
    GB9703369D0 (en) * 1997-02-18 1997-04-09 Lindqvist Bjorn H Process
    JP4086325B2 (en) 1997-04-23 2008-05-14 プリュックテュン,アンドレアス Method for identifying a nucleic acid molecule encoding a (poly) peptide that interacts with a target molecule
    DK1019496T3 (en) * 1997-07-07 2005-01-10 Medical Res Council In vitro sorting method
    US6066460A (en) * 1997-07-24 2000-05-23 President And Fellows Of Harvard College Method for cloning secreted proteins
    US6927025B1 (en) 1997-09-03 2005-08-09 Biovation Limited Methods for protein screening
    ATE311443T1 (en) * 1997-09-03 2005-12-15 Biovation Ltd METHODS FOR SCREENING PROTEINS
    GB9722131D0 (en) 1997-10-20 1997-12-17 Medical Res Council Method
    NZ528767A (en) 1998-03-30 2005-08-26 Northwest Biotherapeutics Inc Therapeutic and diagnostic applications based on the role of the CXCR-4 gene in tumorigenesis
    EP1068356B8 (en) 1998-04-03 2007-01-03 Adnexus Therapeutics, Inc. Addressable protein arrays
    US6440695B1 (en) * 1998-04-17 2002-08-27 Whitehead Institute For Biomedical Research Method for producing diverse libraries of encoded polypeptides
    US6562622B1 (en) * 1998-05-08 2003-05-13 Diatech Pty, Ltd Continuous in vitro evolution
    US20030170820A1 (en) * 1998-05-08 2003-09-11 Gregory Coia Continuous in- vitro evolution
    GB9810756D0 (en) 1998-05-19 1998-07-15 Angeletti P Ist Richerche Bio Mimotopes of hypervariable region 1 of the e2 glycoprotein of hcv and uses thereof
    AU4854999A (en) * 1998-07-02 2000-01-24 Invitro Diagnostics, Inc. Methods, compositions and apparatus for making nucleic acid molecules having a selected affinity to a target molecule
    US6787308B2 (en) * 1998-07-30 2004-09-07 Solexa Ltd. Arrayed biomolecules and their use in sequencing
    US20100130368A1 (en) * 1998-07-30 2010-05-27 Shankar Balasubramanian Method and system for sequencing polynucleotides
    US20040106110A1 (en) * 1998-07-30 2004-06-03 Solexa, Ltd. Preparation of polynucleotide arrays
    US20030022207A1 (en) * 1998-10-16 2003-01-30 Solexa, Ltd. Arrayed polynucleotides and their use in genome analysis
    US20040002101A1 (en) * 1998-08-12 2004-01-01 Proteus S.A. Method and kit for discovering nucleic acids that encode desired functions
    DE69941193D1 (en) 1998-08-17 2009-09-10 Bristol Myers Squibb Co IDENTIFICATION OF COMPOUND PROTEIN INTERACTIONS WITH LIBRARIES OF COUPLED PROTEIN NUCLEIC ACID MOLECULES
    US6312927B1 (en) 1998-08-17 2001-11-06 Phylos, Inc. Methods for producing nucleic acids lacking 3'-untranslated regions and optimizing cellular RNA-protein fusion formation
    US20060088846A1 (en) 1998-08-28 2006-04-27 Michele Pagano Methods to identify compounds useful for the treatment of proliferative and differentiative disorders
    WO2000032823A1 (en) 1998-12-02 2000-06-08 Phylos, Inc. Dna-protein fusions and uses thereof
    US7883704B2 (en) * 1999-03-25 2011-02-08 Abbott Gmbh & Co. Kg Methods for inhibiting the activity of the P40 subunit of human IL-12
    KR101222450B1 (en) 1999-03-25 2013-01-16 애보트 게엠베하 운트 콤파니 카게 Human antibodies that bind human IL-12 and methods for producing
    US6914128B1 (en) 1999-03-25 2005-07-05 Abbott Gmbh & Co. Kg Human antibodies that bind human IL-12 and methods for producing
    DK2385124T3 (en) 1999-05-14 2013-11-18 Arbor Vita Corp Peptides, or peptide analogs, for modulating the binding of a PDZ protein and a PL protein
    EP1187626A4 (en) 1999-06-01 2006-07-19 Compound Therapeutics Inc Methods for producing 5'-nucleic acid-protein conjugates
    US20030091999A1 (en) * 1999-10-01 2003-05-15 Zhongping Yu Compositions and methods for identifying polypeptides and nucleic acid molecules
    US7022479B2 (en) 2000-01-24 2006-04-04 Compound Therapeutics, Inc. Sensitive, multiplexed diagnostic assays for protein analysis
    WO2001053539A1 (en) 2000-01-24 2001-07-26 Phylos, Inc. Sensitive, multiplexed diagnostic assays for protein analysis
    AU3944401A (en) * 2000-03-31 2001-10-15 Cambridge Antibody Tech Improvements to ribosome display
    EP1276849A4 (en) 2000-04-12 2004-06-09 Human Genome Sciences Inc Albumin fusion proteins
    US6902734B2 (en) 2000-08-07 2005-06-07 Centocor, Inc. Anti-IL-12 antibodies and compositions thereof
    UA81743C2 (en) 2000-08-07 2008-02-11 Центокор, Инк. HUMAN MONOCLONAL ANTIBODY WHICH SPECIFICALLY BINDS TUMOR NECROSIS FACTOR ALFA (TNFα), PHARMACEUTICAL MIXTURE CONTAINING THEREOF, AND METHOD FOR TREATING ARTHRITIS
    US7288390B2 (en) 2000-08-07 2007-10-30 Centocor, Inc. Anti-dual integrin antibodies, compositions, methods and uses
    WO2002016412A2 (en) 2000-08-18 2002-02-28 Dyax Corp. Binding polypeptides for b lymphocyte stimulator protein (blys)
    EP1332209B1 (en) 2000-09-08 2009-11-11 Universität Zürich Collections of repeat proteins comprising repeat modules
    JP4723713B2 (en) * 2000-09-25 2011-07-13 トヨタ自動車株式会社 Screening method for potential translational regulator of mRNA
    US20040091509A1 (en) * 2000-12-14 2004-05-13 Avon Products, Inc. Skin care composition that mediates cell to cell communication
    KR100401296B1 (en) * 2000-12-27 2003-10-11 드림바이오젠 주식회사 Protein Muteins Modified By Modifying Materials And Method For Producing The Same
    JP4061043B2 (en) 2000-12-28 2008-03-12 株式会社ポストゲノム研究所 Method for producing peptide etc. by in vitro transcription / translation system
    WO2002074929A2 (en) 2001-03-19 2002-09-26 President And Fellows Of Harvard College Evolving new molecular function
    EP1249499A1 (en) * 2001-04-10 2002-10-16 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Method and device for the determination and selection of molecule-molecule interactions
    DE60236646D1 (en) 2001-04-13 2010-07-22 Human Genome Sciences Inc Anti-VEGF-2 antibodies
    WO2002102820A1 (en) 2001-06-20 2002-12-27 Nuevolution A/S Nucleoside derivatives for library preparation
    ATE371026T1 (en) 2001-06-20 2007-09-15 Nuevolution As A METHOD FOR SYNTHESIS OF MATTRICE-DEPENDENT MOLECULES
    GB0115841D0 (en) 2001-06-28 2001-08-22 Medical Res Council Ligand
    EP1440083B1 (en) 2001-10-25 2013-01-02 Medical Research Council Molecules
    GB0126887D0 (en) * 2001-11-08 2002-01-02 Univ London Method for producing and identifying soluble protein domains
    GB0127564D0 (en) 2001-11-16 2002-01-09 Medical Res Council Emulsion compositions
    WO2003054234A1 (en) * 2001-12-19 2003-07-03 Roche Diagnostics Corporation Pcr based high throughput polypeptide screening
    US20040052928A1 (en) * 2002-09-06 2004-03-18 Ehud Gazit Peptides and methods using same for diagnosing and treating amyloid-associated diseases
    US7781396B2 (en) * 2002-01-31 2010-08-24 Tel Aviv University Future Technology Development L.P. Peptides directed for diagnosis and treatment of amyloid-associated disease
    ATE508188T1 (en) 2002-02-01 2011-05-15 Life Technologies Corp OLIGONUCLEOTIDE COMPOSITIONS WITH IMPROVED EFFECTIVENESS
    US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
    EP1572902B1 (en) 2002-02-01 2014-06-11 Life Technologies Corporation HIGH POTENCY siRNAS FOR REDUCING THE EXPRESSION OF TARGET GENES
    WO2003072054A2 (en) * 2002-02-25 2003-09-04 Cabot Corporation Custom ligand design for biomolecular filtration and purification for bioseperation
    US7413854B2 (en) 2002-03-15 2008-08-19 Nuevolution A/S Method for synthesising templated molecules
    NZ535602A (en) 2002-04-08 2006-07-28 Pioneer Hi Bred Int Enhanced silk exsertion under stress in Zea mays plants
    AU2003231196C1 (en) 2002-05-10 2009-05-21 Henry M. Jackson Foundation For The Advancement Of Military Medicine Androgen-regulated PMEPA1 and cancer
    US20040248094A1 (en) * 2002-06-12 2004-12-09 Ford Lance P. Methods and compositions relating to labeled RNA molecules that reduce gene expression
    US20100075423A1 (en) * 2002-06-12 2010-03-25 Life Technologies Corporation Methods and compositions relating to polypeptides with rnase iii domains that mediate rna interference
    AU2003243541A1 (en) * 2002-06-12 2003-12-31 Ambion, Inc. Methods and compositions relating to labeled rna molecules that reduce gene expression
    US20060002935A1 (en) 2002-06-28 2006-01-05 Domantis Limited Tumor Necrosis Factor Receptor 1 antagonists and methods of use therefor
    US9321832B2 (en) 2002-06-28 2016-04-26 Domantis Limited Ligand
    US10730906B2 (en) 2002-08-01 2020-08-04 Nuevolutions A/S Multi-step synthesis of templated molecules
    JP4657919B2 (en) 2002-08-19 2011-03-23 プレジデント アンド フェローズ オブ ハーバード カレッジ Evolving new molecular functions
    DE60335000D1 (en) 2002-09-06 2010-12-30 Isogenica Ltd IN VITRO PEPTIDE EXPRESSION BANK
    CA2495449A1 (en) 2002-09-09 2004-03-18 Arbor Vita Corporation Methods of diagnosing cervical cancer
    AU2003266932B2 (en) 2002-09-27 2009-04-09 Mpm-Holding Aps Spatially encoded polymer matrix
    EP3299463B1 (en) 2002-10-30 2020-10-21 Nuevolution A/S Enzymatic encoding
    AU2003285151A1 (en) * 2002-11-04 2004-06-07 Bioarctic Neuroscience Ab Methods for the identification of agents that modulate the structure and processing of beta-amyloid precursor protein
    JP2006505271A (en) * 2002-11-04 2006-02-16 アイコジェネックス コーポレイション Methods for the identification of substances that modulate the structure and processing of membrane-bound precursor proteins
    US7491699B2 (en) * 2002-12-09 2009-02-17 Ramot At Tel Aviv University Ltd. Peptide nanostructures and methods of generating and using the same
    WO2004056994A2 (en) 2002-12-19 2004-07-08 Nuevolution A/S Quasirandom structure and function guided synthesis methods
    WO2004060791A1 (en) 2003-01-07 2004-07-22 Ramot At Tel Aviv University Ltd. Peptide nanostructures encapsulating a foreign material and method of manufacturing same
    ES2338654T5 (en) 2003-01-29 2017-12-11 454 Life Sciences Corporation Pearl emulsion nucleic acid amplification
    WO2004074429A2 (en) 2003-02-21 2004-09-02 Nuevolution A/S Method for producing second-generation library
    US10533998B2 (en) 2008-07-18 2020-01-14 Bio-Rad Laboratories, Inc. Enzyme quantification
    DE602004019764D1 (en) 2003-03-20 2009-04-16 Nuevolution As LIGATION-RELATED CODING OF SMALL MOLECULES
    US20060078893A1 (en) 2004-10-12 2006-04-13 Medical Research Council Compartmentalised combinatorial chemistry by microfluidic control
    GB0307428D0 (en) 2003-03-31 2003-05-07 Medical Res Council Compartmentalised combinatorial chemistry
    GB0307403D0 (en) 2003-03-31 2003-05-07 Medical Res Council Selection by compartmentalised screening
    US9708410B2 (en) 2003-05-30 2017-07-18 Janssen Biotech, Inc. Anti-tissue factor antibodies and compositions
    WO2005010145A2 (en) 2003-07-05 2005-02-03 The Johns Hopkins University Method and compositions for detection and enumeration of genetic variations
    US7585815B2 (en) * 2003-07-24 2009-09-08 Lawrence Livermore National Security, Llc High throughput protein production screening
    US20120202710A1 (en) 2003-09-09 2012-08-09 Integrigen, Inc. Methods and compositions for generation of germline human antibody genes
    WO2005026387A1 (en) 2003-09-18 2005-03-24 Nuevolution A/S A method for obtaining structural information concerning an encoded molecule and method for selecting compounds
    CA2540407A1 (en) * 2003-09-25 2005-03-31 Tel Aviv University Future Technology Development L.P. Compositions and methods using same for treating amyloid-associated diseases
    US7625707B2 (en) * 2003-10-02 2009-12-01 Ramot At Tel Aviv University Ltd. Antibacterial agents and methods of identifying and utilizing same
    NZ547723A (en) 2003-12-17 2009-09-25 Praecis Pharm Inc Methods for synthesis of encoded libraries
    US7972994B2 (en) 2003-12-17 2011-07-05 Glaxosmithkline Llc Methods for synthesis of encoded libraries
    EP1709084A4 (en) 2003-12-23 2008-05-28 Nono Inc Polypeptides for modulating binding of trp channel proteins and trp-associated proteins
    EP1730277B1 (en) 2004-03-22 2009-10-28 Nuevolution A/S Ligational encoding using building block oligonucleotides
    US20050221339A1 (en) 2004-03-31 2005-10-06 Medical Research Council Harvard University Compartmentalised screening by microfluidic control
    WO2005108598A1 (en) 2004-05-11 2005-11-17 Axiogenesis Ag Assay for drug discovery based on in vitro differentiated cells
    WO2006006172A2 (en) * 2004-07-15 2006-01-19 Ramot At Tel Aviv University Ltd. Use of anti-amyloid agents for treating and typing pathogen infections
    WO2006013552A2 (en) 2004-08-02 2006-02-09 Ramot At Tel Aviv University Ltd. Articles of peptide nanostructures and method of forming the same
    US7732479B2 (en) 2004-08-19 2010-06-08 Tel Aviv University Future Technology Development L.P. Compositions for treating amyloid associated diseases
    US7335491B2 (en) 2004-08-27 2008-02-26 Wyeth Research Ireland Limited Production of anti-abeta
    TWI384069B (en) 2004-08-27 2013-02-01 Pfizer Ireland Pharmaceuticals Production of polypeptides
    US7300773B2 (en) * 2004-08-27 2007-11-27 Wyeth Research Ireland Limited Production of TNFR-Ig
    US7786086B2 (en) 2004-09-08 2010-08-31 Ramot At Tel-Aviv University Ltd. Peptide nanostructures containing end-capping modified peptides and methods of generating and using the same
    US7968287B2 (en) 2004-10-08 2011-06-28 Medical Research Council Harvard University In vitro evolution in microfluidic systems
    CN101724071A (en) 2004-10-08 2010-06-09 杜门蒂斯有限公司 Single domain antibodies against TNFRl and methods of use therefor
    KR100730875B1 (en) 2004-12-07 2007-06-20 가부시키가이샤 시마즈세이사쿠쇼 DNA fragment to promote translation reaction and method for cell-free system protein synthesis using the same
    US20060142228A1 (en) * 2004-12-23 2006-06-29 Ambion, Inc. Methods and compositions concerning siRNA's as mediators of RNA interference
    JP2008532559A (en) 2005-03-19 2008-08-21 メディカル リサーチ カウンシル Treatment and prevention of viral infection or improvement of treatment and prevention
    JO3058B1 (en) 2005-04-29 2017-03-15 Applied Molecular Evolution Inc Anti-IL-6 Antibodies,Compositions,Methods and uses
    JPWO2006123445A1 (en) * 2005-05-18 2008-12-25 国立大学法人京都大学 Protein display method
    JP2009501006A (en) 2005-06-30 2009-01-15 セントカー・インコーポレーテツド Anti-IL-23 antibodies, compositions, methods and uses
    EP1910828A4 (en) 2005-07-01 2012-03-14 Univ St Louis Phosphospecific chemokine receptor antibodies
    WO2007011722A2 (en) 2005-07-15 2007-01-25 President And Fellows Of Harvard College Reaction discovery system
    US20070111237A1 (en) * 2005-09-14 2007-05-17 Maurizio Zanetti Process for identifying antigenized antibodies using ribosome cell free expression system
    US10004828B2 (en) * 2005-10-11 2018-06-26 Romat at Tel-Aviv University Ltd. Self-assembled Fmoc-ff hydrogels
    EP1940755A2 (en) 2005-10-28 2008-07-09 Praecis Pharmaceuticals Inc. Methods for identifying compounds of interest using encoded libraries
    US7879212B2 (en) * 2005-11-03 2011-02-01 Ramot At Tel-Aviv University Ltd. Peptide nanostructure-coated electrodes
    LT3305900T (en) 2005-12-01 2021-11-10 Nuevolution A/S Enzymatic encoding methods for efficient synthesis of large libraries
    HUE034269T2 (en) 2005-12-29 2018-02-28 Janssen Biotech Inc Human anti-il-23 antibodies, compositions, methods and uses
    WO2007081386A2 (en) 2006-01-11 2007-07-19 Raindance Technologies, Inc. Microfluidic devices and methods of use
    EP2010569A4 (en) 2006-03-20 2009-09-09 Xoma Technology Ltd Human antibodies specific for gastrin materials and methods
    US7749957B2 (en) 2006-04-06 2010-07-06 E.I. Du Pont De Nemours And Company Clay-binding peptides and methods of use
    US9562837B2 (en) 2006-05-11 2017-02-07 Raindance Technologies, Inc. Systems for handling microfludic droplets
    US20080014589A1 (en) 2006-05-11 2008-01-17 Link Darren R Microfluidic devices and methods of use thereof
    WO2008005290A2 (en) 2006-06-29 2008-01-10 The Trustees Of Columbia University In The City Of New York Methods for testing anti-thrombotic agents
    DK2043671T3 (en) 2006-07-11 2020-06-15 Nono Inc PEPTID AND COMPOSITION THEREOF USED FOR TREATMENT OF FEBRUARY CASES
    EP2077912B1 (en) 2006-08-07 2019-03-27 The President and Fellows of Harvard College Fluorocarbon emulsion stabilizing surfactants
    US9580515B2 (en) 2006-08-21 2017-02-28 Zensun (Shanghai) Science & Technology, Co., Ltd. Neukinase, a downstream protein of neuregulin
    WO2008063933A2 (en) 2006-11-10 2008-05-29 Massachusetts Institute Of Technology Pak modulators
    RU2554747C9 (en) 2006-12-20 2015-10-20 Ксома (Сша) Ллс Method of treating il-1beta-dependent diseases
    NZ578065A (en) 2007-01-16 2012-09-28 Abbott Lab Methods for treating psoriasis with an antibody which binds to an epitope
    US8772046B2 (en) 2007-02-06 2014-07-08 Brandeis University Manipulation of fluids and reactions in microfluidic systems
    NZ598881A (en) * 2007-03-29 2013-11-29 Abbvie Inc Crystalline anti-human il-12 antibodies
    US8592221B2 (en) 2007-04-19 2013-11-26 Brandeis University Manipulation of fluids, fluid components and reactions in microfluidic systems
    ES2614735T3 (en) 2007-07-23 2017-06-01 Janssen Biotech, Inc. Methods and compositions for treating fibrosis-related disorders using IL-17 antagonists
    US7951559B2 (en) * 2007-07-25 2011-05-31 E.I. Du Pont De Nemours And Company Recombinant peptide production using a cross-linkable solubility tag
    US7678883B2 (en) * 2007-07-25 2010-03-16 E.I. Du Pont De Nemours And Company Solubility tags for the expression and purification of bioactive peptides
    US7829311B2 (en) 2007-07-25 2010-11-09 E.I. Du Pont De Nemours And Company Ketosteroid isomerase inclusion body tag engineered to be acid-resistant by replacing aspartates with glutamate
    US7794963B2 (en) 2007-11-02 2010-09-14 E.I. Du Pont De Nemours And Company Use of tetracysteine tags in fluorescence-activated cell sorting analysis of prokaryotic cells producing peptides or proteins
    BRPI0819743A2 (en) 2007-11-20 2014-10-07 Pioneer Hi Bred Int ISOLATED NUCLEIC ACID, EXPRESSION CASSETTE, HOST CELL, TRANSGENIC PLANT, TRANSGENIC SEED, METHOD FOR MODULATING ETHYLEN RESPONSE ON A PLANT, PROTEIN ISOLATED
    PT2391650E (en) 2007-12-20 2015-01-14 Xoma Us Llc Methods for the treatment of gout
    NZ587765A (en) 2008-03-18 2013-02-22 Abbott Lab Methods for treating psoriasis
    GB0806562D0 (en) 2008-04-10 2008-05-14 Fermentas Uab Production of nucleic acid
    SG176464A1 (en) 2008-05-09 2011-12-29 Agency Science Tech & Res Diagnosis and treatment of kawasaki disease
    WO2010009365A1 (en) 2008-07-18 2010-01-21 Raindance Technologies, Inc. Droplet libraries
    EP2316030B1 (en) 2008-07-25 2019-08-21 Wagner, Richard W. Protein screeing methods
    US8563697B2 (en) 2008-08-14 2013-10-22 Cephalon Australia Pty. Ltd. Anti-IL-12/IL-23 antibodies
    EP2364359A2 (en) * 2008-09-26 2011-09-14 Wyeth LLC Compatible display vector systems
    US20100113304A1 (en) * 2008-09-26 2010-05-06 Wyeth Compatible display vector systems
    CA2741834C (en) 2008-10-31 2022-04-05 Centocor Ortho Biotech Inc. Fibronectin type iii domain based scaffold compositions, methods and uses
    DK2356270T3 (en) 2008-11-07 2016-12-12 Fabrus Llc Combinatorial antibody libraries and uses thereof
    WO2010062896A1 (en) * 2008-11-28 2010-06-03 Abbott Laboratories Stable antibody compositions and methods for stabilizing same
    US8287845B2 (en) * 2008-12-18 2012-10-16 E I Du Pont De Nemours And Company Hair-binding peptides
    US20100158846A1 (en) * 2008-12-18 2010-06-24 E. I. Du Pont De Nemours And Company Hair-binding peptides
    US20100158822A1 (en) 2008-12-18 2010-06-24 E .I. Du Pont De Nemours And Company Peptides that bind to silica-coated particles
    US20100158837A1 (en) 2008-12-18 2010-06-24 E. I. Du Pont De Nemours And Company Iron oxide-binding peptides
    JP2012513464A (en) 2008-12-23 2012-06-14 ザ トラスティーズ オブ コロンビア ユニヴァーシティ イン ザ シティ オブ ニューヨーク Phosphodiesterase inhibitors and uses thereof
    WO2010074783A1 (en) 2008-12-23 2010-07-01 The Trustees Of Columbia University In The City Of New York Phosphodiesterase inhibitors and uses thereof
    EP2396011B1 (en) 2009-02-12 2016-04-13 Janssen Biotech, Inc. Fibronectin type iii domain based scaffold compositions, methods and uses
    WO2010101818A1 (en) 2009-03-02 2010-09-10 Pioneer Hi-Bred International, Inc. Nac transcriptional activators involved in abiotic stress tolerance
    US8528589B2 (en) 2009-03-23 2013-09-10 Raindance Technologies, Inc. Manipulation of microfluidic droplets
    US8481678B2 (en) 2009-03-30 2013-07-09 E I Du Pont De Nemours And Company Peptide-based tooth whitening reagents
    JP2012522057A (en) * 2009-03-30 2012-09-20 ジヨンソン・アンド・ジヨンソン・コンシユーマー・カンパニーズ・インコーポレーテツド Peptide-based systems for the delivery of cosmetic agents
    US20110035843A1 (en) 2009-08-05 2011-02-10 Pioneer Hi-Bred International, Inc. Novel eto1 genes and use of same for reduced ethylene and improved stress tolerance in plants
    KR101161622B1 (en) * 2009-08-31 2012-07-04 헬릭스 주식회사 DNA fragment to promote translation efficiency and recombinant vectors containing the same
    KR20140048229A (en) * 2009-09-14 2014-04-23 애브비 인코포레이티드 Methods for treating psoriasis
    US9885711B2 (en) 2009-09-25 2018-02-06 Xoma Technology Ltd. Screening methods
    US8926976B2 (en) 2009-09-25 2015-01-06 Xoma Technology Ltd. Modulators
    EP2486409A1 (en) 2009-10-09 2012-08-15 Universite De Strasbourg Labelled silica-based nanomaterial with enhanced properties and uses thereof
    DK2488867T3 (en) * 2009-10-14 2020-11-09 Janssen Biotech Inc PROCEDURES FOR AFFINITY MATURE OF ANTIBODIES
    US20110165648A1 (en) 2009-11-04 2011-07-07 Menno Van Lookeren Campagne Co-crystal structure of factor D and anti-factor D antibody
    IL201999A (en) 2009-11-08 2017-10-31 Medical Res & Development Fund For Health Services Bnai Zion Medical Center The State Of Israel Knock-out mouse of the mo-1 gene, a gene associated with morbid obesity
    US10640457B2 (en) 2009-12-10 2020-05-05 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase activators and uses thereof
    WO2011072243A1 (en) 2009-12-10 2011-06-16 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase activators and uses thereof
    US10837883B2 (en) 2009-12-23 2020-11-17 Bio-Rad Laboratories, Inc. Microfluidic systems and methods for reducing the exchange of molecules between droplets
    CN102933231B (en) 2010-02-10 2015-07-29 伊缪诺金公司 CD20 antibody and uses thereof
    US9366632B2 (en) 2010-02-12 2016-06-14 Raindance Technologies, Inc. Digital analyte analysis
    WO2011100604A2 (en) 2010-02-12 2011-08-18 Raindance Technologies, Inc. Digital analyte analysis
    US9399797B2 (en) 2010-02-12 2016-07-26 Raindance Technologies, Inc. Digital analyte analysis
    US10351905B2 (en) 2010-02-12 2019-07-16 Bio-Rad Laboratories, Inc. Digital analyte analysis
    JP5863045B2 (en) 2010-03-11 2016-02-16 国立研究開発法人理化学研究所 Method for selecting polypeptide sequence, metal oxide or silicon-containing compound binding peptide and use thereof
    US11225655B2 (en) 2010-04-16 2022-01-18 Nuevolution A/S Bi-functional complexes and methods for making and using such complexes
    AU2011245225B2 (en) 2010-04-30 2015-09-17 Janssen Biotech, Inc. Stabilized fibronectin domain compositions, methods and uses
    WO2012022734A2 (en) 2010-08-16 2012-02-23 Medimmune Limited Anti-icam-1 antibodies and methods of use
    WO2012023085A1 (en) 2010-08-20 2012-02-23 Wyeth Llc Cell culture of growth factor-free adapted cells
    US8987415B2 (en) 2010-09-07 2015-03-24 Technische Universitaet Muenchen Recombinant bacterial lipocalin blc and uses thereof
    EP3447155A1 (en) 2010-09-30 2019-02-27 Raindance Technologies, Inc. Sandwich assays in droplets
    US9198911B2 (en) 2010-11-02 2015-12-01 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
    HUE045869T2 (en) 2010-11-02 2020-01-28 Univ Columbia Methods for treating hair loss disorders
    CA2822621C (en) 2010-12-22 2020-12-15 The Trustees Of Columbia University In The City Of New York Histone acetyltransferase modulators and uses thereof
    DE102010056289A1 (en) 2010-12-24 2012-06-28 Geneart Ag Process for the preparation of reading frame correct fragment libraries
    WO2012109600A2 (en) 2011-02-11 2012-08-16 Raindance Technologies, Inc. Methods for forming mixed droplets
    WO2012112804A1 (en) 2011-02-18 2012-08-23 Raindance Technoligies, Inc. Compositions and methods for molecular labeling
    JP5948673B2 (en) 2011-03-10 2016-07-06 ジーンフロンティア株式会社 COMPOSITION FOR PROTEIN SYNTHESIS WITH REDUCED LIPO POLUCHIsaccharide CONTENT AND METHOD FOR PRODUCING PROTEIN USING THE COMPOSITION
    EP2691101A2 (en) 2011-03-31 2014-02-05 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
    US20120301904A1 (en) * 2011-04-26 2012-11-29 Prosetta Antiviral, Inc Multiprotein assemblies
    WO2012161227A1 (en) 2011-05-23 2012-11-29 独立行政法人理化学研究所 Nucleic acid construct, nucleic acid-protein complex, and use thereof
    US8841071B2 (en) 2011-06-02 2014-09-23 Raindance Technologies, Inc. Sample multiplexing
    SG10201902706VA (en) 2011-06-03 2019-04-29 Xoma Technology Ltd Antibodies specific for tgf-beta
    US8658430B2 (en) 2011-07-20 2014-02-25 Raindance Technologies, Inc. Manipulating droplet size
    EP2750767A4 (en) 2011-09-02 2015-10-14 Univ Columbia CaMKII, IP3R, CALCINEURIN, P38 AND MK2/3 INHIBITORS TO TREAT METABOLIC DISTURBANCES OF OBESITY
    SG10201912964PA (en) 2011-09-19 2020-02-27 Axon Neuroscience Se Protein-based therapy and diagnosis of tau-mediated pathology in alzheimer's disease
    KR102035713B1 (en) 2011-09-27 2019-10-24 얀센 바이오테크 인코포레이티드 Fibronectin type iii repeat based protein scaffolds with alternative binding surfaces
    ES2784146T3 (en) 2011-10-21 2020-09-22 Pfizer Addition of iron to improve cell culture
    BR112014009968A2 (en) 2011-10-25 2017-07-04 Du Pont method for reducing acetate, arabinosidase and / or ferulate content in a plant, method of modulating plant carbohydrate concentration in a transgenic plant, method for changing acetyl content and cross-linking in plant tissues, method for production of biomass for biofuel or silage production, product, plant fodder
    US20150017157A1 (en) 2011-12-19 2015-01-15 Xoma (Us) Llc Methods for treating acne
    PL2830662T3 (en) 2012-03-29 2019-02-28 The Trustees Of Columbia University In The City Of New York Methods for treating hair loss disorders
    US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
    CA2868398A1 (en) 2012-04-02 2013-10-10 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
    EP2657334B1 (en) 2012-04-26 2016-07-06 GeneFrontier Corporation Efficient method for displaying protein multimer
    CA2873511A1 (en) 2012-05-17 2013-11-21 Ra Pharmaceuticals, Inc. Peptide and peptidomimetic inhibitors
    GB201214029D0 (en) * 2012-08-07 2012-09-19 Queen Mary & Westfield College method
    KR20240005211A (en) 2012-11-21 2024-01-11 얀센 바이오테크 인코포레이티드 BISPECIFIC EGFR/c-Met ANTIBODIES
    US9695228B2 (en) 2012-11-21 2017-07-04 Janssen Biotech, Inc. EGFR and c-Met fibronectin type III domain binding molecules
    US11377470B2 (en) 2013-03-15 2022-07-05 Modernatx, Inc. Ribonucleic acid purification
    US10138507B2 (en) 2013-03-15 2018-11-27 Modernatx, Inc. Manufacturing methods for production of RNA transcripts
    WO2014152507A2 (en) 2013-03-15 2014-09-25 Pioneer Hi-Bred International, Inc. Modulation of acc deaminase expression
    EP2983804A4 (en) 2013-03-15 2017-03-01 Moderna Therapeutics, Inc. Ion exchange purification of mrna
    US10077439B2 (en) 2013-03-15 2018-09-18 Modernatx, Inc. Removal of DNA fragments in mRNA production process
    DK3019619T3 (en) 2013-07-11 2021-10-11 Modernatx Inc COMPOSITIONS INCLUDING SYNTHETIC POLYNUCLEOTIDES CODING CRISPR-RELATED PROTEINS, SYNTHETIC SGRNAs, AND USES OF USE
    US10385088B2 (en) 2013-10-02 2019-08-20 Modernatx, Inc. Polynucleotide molecules and uses thereof
    US11901041B2 (en) 2013-10-04 2024-02-13 Bio-Rad Laboratories, Inc. Digital analysis of nucleic acid modification
    AU2014334627B2 (en) 2013-10-14 2019-07-25 Janssen Biotech, Inc. Cysteine engineered fibronectin type III domain binding molecules
    US9309314B2 (en) 2013-12-03 2016-04-12 Agency For Science, Technology And Research (A*Star) Polypeptides, nucleic acids and uses thereof
    US9944977B2 (en) 2013-12-12 2018-04-17 Raindance Technologies, Inc. Distinguishing rare variations in a nucleic acid sequence from a sample
    PL3712174T3 (en) 2013-12-24 2022-07-04 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments
    EP3090063B1 (en) 2013-12-31 2019-11-06 Bio-Rad Laboratories, Inc. Method for detection of latent retrovirus
    EP3119876A1 (en) 2014-03-19 2017-01-25 Pfizer Inc. Method of cell culture
    PT3628680T (en) 2014-06-12 2021-10-07 Ra Pharmaceuticals Inc Modulation of complement activity
    WO2015196128A2 (en) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
    US10562946B2 (en) 2014-06-20 2020-02-18 Genentech, Inc. Chagasin-based scaffold compositions, methods, and uses
    EP3169335B8 (en) 2014-07-16 2019-10-09 ModernaTX, Inc. Circular polynucleotides
    JP6640229B2 (en) 2015-01-28 2020-02-05 ラ ファーマシューティカルズ インコーポレイテッドRa Pharmaceuticals,Inc. Modulators of complement activity
    EP3265491A1 (en) 2015-03-03 2018-01-10 Xoma (Us) Llc Treatment of post-prandial hyperinsulinemia and hypoglycemia after bariatric surgery
    WO2016161410A2 (en) 2015-04-03 2016-10-06 Xoma Technology Ltd. Treatment of cancer using inhibitors of tgf-beta and pd-1
    CN107787332B (en) 2015-04-24 2022-09-09 豪夫迈·罗氏有限公司 Multispecific antigen binding proteins
    JP7366518B2 (en) 2015-05-06 2023-10-23 ヤンセン バイオテツク,インコーポレーテツド Prostate-specific membrane antigen-binding fibronectin type III domain
    JP7026509B2 (en) 2015-06-24 2022-02-28 ヤンセン ファーマシューティカ エヌブイ Anti-VISTA antibody and fragment
    CA2994841A1 (en) 2015-08-06 2017-02-09 Xoma (Us) Llc Antibody fragments against the insulin receptor and uses thereof to treat hypoglycemia
    US10647981B1 (en) 2015-09-08 2020-05-12 Bio-Rad Laboratories, Inc. Nucleic acid library generation methods and compositions
    US11434486B2 (en) 2015-09-17 2022-09-06 Modernatx, Inc. Polynucleotides containing a morpholino linker
    WO2017051347A2 (en) 2015-09-23 2017-03-30 Pfizer Inc. Cells and method of cell culture
    MX2018007352A (en) 2015-12-16 2019-05-16 Ra Pharmaceuticals Inc Modulators of complement activity.
    CN116920085A (en) 2016-02-12 2023-10-24 詹森药业有限公司 anti-VISTA (B7H 5) antibodies
    AU2017241776A1 (en) 2016-03-29 2018-10-11 Janssen Biotech, Inc. Treating psoriasis with increased interval dosing of anti-IL12 and/or -23 antibody
    SG11201808356UA (en) 2016-04-05 2018-10-30 Pfizer Cell culture process
    WO2017175058A1 (en) 2016-04-07 2017-10-12 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
    UA126021C2 (en) 2016-06-03 2022-08-03 Янссен Байотек, Інк. Serum albumin-binding fibronectin type iii domains
    EP3471750A4 (en) 2016-06-21 2020-02-26 Janssen Biotech, Inc. Cysteine engineered fibronectin type iii domain binding molecules
    CA3028002A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Method of identifying peptide epitopes, molecules that bind such epitopes and related uses
    MA45491A (en) 2016-06-27 2019-05-01 Juno Therapeutics Inc CMH-E RESTRICTED EPITOPES, BINDING MOLECULES AND RELATED METHODS AND USES
    US20190177421A1 (en) 2016-07-15 2019-06-13 Poseida Therapeutics, Inc. Chimeric antigen receptors and methods for use
    EP3484927A1 (en) 2016-07-15 2019-05-22 Poseida Therapeutics, Inc. Chimeric antigen receptors (cars) specific for muc1 and methods for their use
    EP3490600A1 (en) 2016-08-01 2019-06-05 Xoma (Us) Llc Parathyroid hormone receptor 1 (pth1r) antibodies and uses thereof
    BR112019004711A2 (en) 2016-09-14 2019-05-28 Janssen Biotech Inc chimeric antigen receptors comprising bcma specific type III fibronectin domains and uses thereof
    CA3038679A1 (en) 2016-09-28 2018-04-05 Xoma (Us) Llc Antibodies that bind interleukin-2 and uses thereof
    MX2019003703A (en) 2016-09-30 2020-08-13 Janssen Biotech Inc Safe and effective method of treating psoriasis with anti-il23 specific antibody.
    AU2017362222A1 (en) 2016-11-16 2019-05-30 Janssen Biotech, Inc. Method of treating psoriasis with anti-IL-23 specific antibody
    MX2019006527A (en) 2016-12-07 2019-08-01 Ra Pharmaceuticals Inc Modulators of complement activity.
    CA3046963A1 (en) 2016-12-14 2018-06-21 Janssen Biotech, Inc. Cd8a-binding fibronectin type iii domains
    US10597438B2 (en) 2016-12-14 2020-03-24 Janssen Biotech, Inc. PD-L1 binding fibronectin type III domains
    EP3554561B1 (en) 2016-12-14 2023-06-28 Janssen Biotech, Inc. Cd137 binding fibronectin type iii domains
    EP3354736B1 (en) * 2017-01-25 2020-07-01 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Promoter construct for cell-free protein synthesis
    CA3052095A1 (en) 2017-01-30 2018-08-02 Janssen Biotech, Inc. Anti-tnf antibodies, compositions, and methods for the treatment of active psoriatic arthritis
    MA47442A (en) 2017-02-07 2019-12-18 Janssen Biotech Inc ANTI-TNF ANTIBODIES, COMPOSITIONS AND METHODS FOR THE TREATMENT OF ACTIVE ANKYLOSING SPONDYLARTHRITIS
    WO2018169948A1 (en) 2017-03-13 2018-09-20 Poseida Therapeutics, Inc. Compositions and methods for selective elimination and replacement of hematopoietic stem cells
    WO2018167621A1 (en) 2017-03-16 2018-09-20 Pfizer Inc. Tyrosine prototrophy
    JP2020532987A (en) 2017-09-08 2020-11-19 ポセイダ セラピューティクス,インコーポレイティド Compositions and Methods for Chimeric Ligand Receptor (CLR) -mediated Conditional Gene Expression
    EP3681989A1 (en) 2017-09-15 2020-07-22 Bristol-Myers Squibb Company Online biomass capacitance monitoring during large scale production of polypeptides of interest
    TW201922780A (en) 2017-09-25 2019-06-16 美商健生生物科技公司 Safe and effective method of treating Lupus with anti-IL12/IL23 antibody
    WO2019150309A1 (en) 2018-02-02 2019-08-08 Hammack Scott Modulators of gpr68 and uses thereof for treating and preventing diseases
    JP2021515770A (en) 2018-03-05 2021-06-24 ヤンセン バイオテツク,インコーポレーテツド Treatment of Crohn's disease using anti-IL-23 specific antibody
    KR20200133240A (en) 2018-03-16 2020-11-26 브리스톨-마이어스 스큅 컴퍼니 Metabolic enzyme activity and disulfide bond reduction during protein production
    US20190345245A1 (en) 2018-05-11 2019-11-14 Janssen Biotech, Inc. Methods of Treating Crohn's Disease with Anti-IL23 Specific Antibody
    WO2020016838A2 (en) 2018-07-18 2020-01-23 Janssen Biotech, Inc. Sustained response predictors after treatment with anti-il23 specific antibody
    DK3883606T5 (en) 2018-09-24 2024-01-02 Janssen Biotech Inc SAFE AND EFFECTIVE METHOD FOR TREATMENT OF ULCERATIVE COLITIS WITH ANTI-IL12/IL23 ANTIBODY
    WO2020104943A2 (en) 2018-11-20 2020-05-28 Janssen Biotech, Inc. Safe and effective method of treating psoriasis with anti-il-23 specific antibody
    CA3121884A1 (en) 2018-12-06 2020-06-11 Pfizer Inc. Cells with reduced inhibitor production and methods of use thereof
    US20200197517A1 (en) 2018-12-18 2020-06-25 Janssen Biotech, Inc. Safe and Effective Method of Treating Lupus with Anti-IL12/IL23 Antibody
    WO2020132396A1 (en) 2018-12-20 2020-06-25 Poseida Therapeutics, Inc. Nanotransposon compositions and methods of use
    BR112021013903A2 (en) 2019-01-15 2021-09-21 Janssen Biotech, Inc. COMPOSITIONS AND METHODS OF ANTI-TNF ANTIBODIES FOR THE TREATMENT OF JUVENILE IDIOPATHIC ARTHRITIS
    WO2020152544A1 (en) 2019-01-23 2020-07-30 Janssen Biotech, Inc. Anti-tnf antibody compositions for use in methods for the treatment of psoriatic arthritis
    SG11202107720UA (en) 2019-01-31 2021-08-30 Agency Science Tech & Res Cnx/erp57 inhibitor for use in the treatment or prevention of cancer
    CN113840838A (en) 2019-03-14 2021-12-24 詹森生物科技公司 Methods of manufacture of compositions for the production of anti-TNF antibodies
    EP3938391A1 (en) 2019-03-14 2022-01-19 Janssen Biotech, Inc. Methods for producing anti-tnf antibody compositions
    JP2022525145A (en) 2019-03-14 2022-05-11 ヤンセン バイオテツク,インコーポレーテツド A production method for producing an anti-IL12 / IL23 antibody composition.
    EP3938392A1 (en) 2019-03-14 2022-01-19 Janssen Biotech, Inc. Methods for producing anti-tnf antibody compositions
    KR20210141583A (en) 2019-03-18 2021-11-23 얀센 바이오테크 인코포레이티드 Methods of Treating Psoriasis in Children Using Anti-IL-12/IL-23 Antibodies
    KR20220005568A (en) 2019-05-09 2022-01-13 제넨테크, 인크. Methods for making antibodies
    JP2022534020A (en) 2019-05-23 2022-07-27 ヤンセン バイオテツク,インコーポレーテツド Methods of treating inflammatory bowel disease with combination therapy of antibodies against IL-23 and TNF-alpha
    MX2021014882A (en) 2019-06-03 2022-03-25 Janssen Biotech Inc Anti-tnf antibodies, compositions, and methods for the treatment of active ankylosing spondylitis.
    EP3976648A1 (en) 2019-06-03 2022-04-06 Janssen Biotech, Inc. Anti-tnf antibody compositions, and methods for the treatment of psoriatic arthritis
    WO2021028752A1 (en) 2019-08-15 2021-02-18 Janssen Biotech, Inc. Anti-tfn antibodies for treating type i diabetes
    CN114761424A (en) 2019-09-05 2022-07-15 波赛达治疗公司 Allogeneic cell compositions and methods of use
    WO2021076574A2 (en) 2019-10-14 2021-04-22 Aro Biotherapeutics Company Fn3 domain-sirna conjugates and uses thereof
    CN114786682A (en) 2019-10-14 2022-07-22 Aro生物疗法公司 Fibronectin type III domain of CD71
    AU2020407235A1 (en) 2019-12-20 2022-07-07 Poseida Therapeutics, Inc. Anti-MUC1 compositions and methods of use
    WO2021183795A1 (en) 2020-03-11 2021-09-16 Poseida Therapeutics, Inc. Chimeric stimulatory receptors and methods of use in t cell activation and differentiation
    IL297025A (en) 2020-04-14 2022-12-01 Poseida Therapeutics Inc Compositions and methods for use in the treatment of cancer
    US20240000969A1 (en) 2020-10-21 2024-01-04 Poseida Therapeutics San Diego Compositions and methods for delivery of nucleic acids
    EP4305062A1 (en) 2021-03-12 2024-01-17 Janssen Biotech, Inc. Method of treating psoriatic arthritis patients with inadequate response to tnf therapy with anti-il23 specific antibody
    IL305802A (en) 2021-03-12 2023-11-01 Janssen Biotech Inc Safe and effective method of treating psoriatic arthritis with anti-il23 specific antibody
    AU2022306973A1 (en) 2021-07-09 2024-02-22 Janssen Biotech, Inc. Manufacturing methods for producing anti-il12/il23 antibody compositions
    US20230042465A1 (en) 2021-07-09 2023-02-09 Janssen Biotech, Inc. Manufacturing Methods for Producing Anti-TNF Antibody Compositions
    AU2022306144A1 (en) 2021-07-09 2024-02-22 Janssen Biotech, Inc. Manufacturing methods for producing anti-tnf antibody compositions
    AU2022360244A1 (en) 2021-10-04 2024-04-11 Poseida Therapeutics, Inc. Transposon compositions and methods of use thereof
    WO2023073615A1 (en) 2021-10-29 2023-05-04 Janssen Biotech, Inc. Methods of treating crohn's disease with anti-il23 specific antibody
    WO2023079058A1 (en) 2021-11-05 2023-05-11 Yokogawa Insilico Biotechnology Gmbh Cell culture with reduced production of lactate
    WO2023084488A1 (en) 2021-11-15 2023-05-19 Janssen Biotech, Inc. Methods of treating crohn's disease with anti-il23 specific antibody
    US20230159633A1 (en) 2021-11-23 2023-05-25 Janssen Biotech, Inc. Method of Treating Ulcerative Colitis with Anti-IL23 Specific Antibody
    WO2023141576A1 (en) 2022-01-21 2023-07-27 Poseida Therapeutics, Inc. Compositions and methods for delivery of nucleic acids
    WO2023148598A1 (en) 2022-02-02 2023-08-10 Pfizer Inc. Cysteine prototrophy
    WO2023187707A1 (en) 2022-03-30 2023-10-05 Janssen Biotech, Inc. Method of treating mild to moderate psoriasis with il-23 specific antibody
    WO2023223265A1 (en) 2022-05-18 2023-11-23 Janssen Biotech, Inc. Method for evaluating and treating psoriatic arthritis with il23 antibody

    Citations (1)

    * Cited by examiner, † Cited by third party
    Publication number Priority date Publication date Assignee Title
    GB2183661A (en) * 1985-03-30 1987-06-10 Marc Ballivet Method for obtaining dna, rna, peptides, polypeptides or proteins by means of a dna recombinant technique

    Family Cites Families (5)

    * Cited by examiner, † Cited by third party
    Publication number Priority date Publication date Assignee Title
    US4710464A (en) * 1984-09-27 1987-12-01 Eli Lilly And Company Transcription terminators
    ZA862334B (en) * 1985-03-28 1987-11-25 Cetus Corp Process for amplifying,detecting and/or cloning nucleic acid sequences
    US4683202A (en) * 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
    US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
    US4800159A (en) * 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences

    Patent Citations (1)

    * Cited by examiner, † Cited by third party
    Publication number Priority date Publication date Assignee Title
    GB2183661A (en) * 1985-03-30 1987-06-10 Marc Ballivet Method for obtaining dna, rna, peptides, polypeptides or proteins by means of a dna recombinant technique

    Non-Patent Citations (1)

    * Cited by examiner, † Cited by third party
    Title
    Methods in Enzymology, vol. 152, 1987, pages 393-399. *

    Also Published As

    Publication number Publication date
    DE69032483D1 (en) 1998-08-20
    KR920703839A (en) 1992-12-18
    US5658754A (en) 1997-08-19
    EP0494955A4 (en) 1992-08-12
    ES2118066T3 (en) 1998-09-16
    DE69032483T2 (en) 1998-11-26
    ATE168416T1 (en) 1998-08-15
    JPH05503000A (en) 1993-05-27
    AU6537390A (en) 1991-04-28
    KR100204359B1 (en) 1999-06-15
    WO1991005058A1 (en) 1991-04-18
    CA2067194A1 (en) 1991-04-06
    JP3127158B2 (en) 2001-01-22
    KR100204360B1 (en) 1999-06-15
    EP0494955A1 (en) 1992-07-22
    US5643768A (en) 1997-07-01
    JP2001078787A (en) 2001-03-27
    DK0494955T3 (en) 1998-10-26
    CA2067194C (en) 2003-03-18
    AU638762B2 (en) 1993-07-08
    KR0185192B1 (en) 1999-04-01

    Similar Documents

    Publication Publication Date Title
    EP0494955B1 (en) Cell-free synthesis and isolation of novel genes and polypeptides
    US5843701A (en) Systematic polypeptide evolution by reverse translation
    US8137906B2 (en) Method for the synthesis of DNA fragments
    EP1151143B1 (en) Selection of proteins using rna-protein fusions
    US5922545A (en) In vitro peptide and antibody display libraries
    US6107059A (en) Peptide library and screening method
    EP1712623B1 (en) Selection of proteins using RNA-protein fusions
    CN100587068C (en) Use of multiple recombination sites with unique specificity in recombinational cloning
    WO1993003172A1 (en) Systematic polypeptide evolution by reverse translation
    WO1992002536A1 (en) Systematic polypeptide evolution by reverse translation
    WO1995011922A1 (en) In vitro peptide and antibody display libraries
    JP4519318B2 (en) Continuous in vitro evolution
    JPH0856667A (en) Method of obtaining dna,rna,peptide,polypeptide or protein by recombinant dna technology
    WO2007136840A2 (en) Nucleic acid library design and assembly
    US20030124537A1 (en) Procaryotic libraries and uses
    WO2021190629A1 (en) Construction method and application of antigen-specific binding polypeptide gene display vector
    WO2002008408A2 (en) Modular vector systems
    CN1399684A (en) Methods of manipulating and sequencing nucleic acid molecules using transposition and recombination
    US20090176281A1 (en) Modular vector systems
    US6265545B1 (en) Reading frame independent epitope tagging
    US20020038000A1 (en) Systematic polypeptide evolution by reverse translation
    US20070048774A1 (en) Continuous in-vitro evolution
    EP0628076A1 (en) PRODUCTION OF MONOCLONAL RECOMBINANT ANTIBODIES WITHOUT THE USE OF HYBRIDOMAS BY $i(IN VITRO) SPLEEN FRAGMENT CULTURE COMBINED WITH ISOTHERMAL SELF-SUSTAINED SEQUENCE REPLICATION OF RNA
    JP2002510502A (en) Method for producing a library of expressible gene sequences
    WO2004001036A1 (en) Compositions and methods for selecting open reading frames

    Legal Events

    Date Code Title Description
    PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

    Free format text: ORIGINAL CODE: 0009012

    17P Request for examination filed

    Effective date: 19920403

    AK Designated contracting states

    Kind code of ref document: A1

    Designated state(s): AT BE CH DE DK ES FR GB IT LI LU NL SE

    A4 Supplementary search report drawn up and despatched

    Effective date: 19920619

    AK Designated contracting states

    Kind code of ref document: A4

    Designated state(s): AT BE CH DE DK ES FR GB IT LI LU NL SE

    RAP1 Party data changed (applicant data changed or rights of an application transferred)

    Owner name: OPTEIN, INC.

    RIN1 Information on inventor provided before grant (corrected)

    Inventor name: KAWASAKI, GLENN

    17Q First examination report despatched

    Effective date: 19941229

    GRAG Despatch of communication of intention to grant

    Free format text: ORIGINAL CODE: EPIDOS AGRA

    GRAG Despatch of communication of intention to grant

    Free format text: ORIGINAL CODE: EPIDOS AGRA

    GRAG Despatch of communication of intention to grant

    Free format text: ORIGINAL CODE: EPIDOS AGRA

    GRAH Despatch of communication of intention to grant a patent

    Free format text: ORIGINAL CODE: EPIDOS IGRA

    GRAH Despatch of communication of intention to grant a patent

    Free format text: ORIGINAL CODE: EPIDOS IGRA

    GRAA (expected) grant

    Free format text: ORIGINAL CODE: 0009210

    AK Designated contracting states

    Kind code of ref document: B1

    Designated state(s): AT BE CH DE DK ES FR GB IT LI LU NL SE

    REF Corresponds to:

    Ref document number: 168416

    Country of ref document: AT

    Date of ref document: 19980815

    Kind code of ref document: T

    REG Reference to a national code

    Ref country code: CH

    Ref legal event code: EP

    ITF It: translation for a ep patent filed

    Owner name: JACOBACCI & PERANI S.P.A.

    REF Corresponds to:

    Ref document number: 69032483

    Country of ref document: DE

    Date of ref document: 19980820

    ET Fr: translation filed
    REG Reference to a national code

    Ref country code: ES

    Ref legal event code: FG2A

    Ref document number: 2118066

    Country of ref document: ES

    Kind code of ref document: T3

    REG Reference to a national code

    Ref country code: CH

    Ref legal event code: NV

    Representative=s name: BOVARD AG PATENTANWAELTE

    REG Reference to a national code

    Ref country code: DK

    Ref legal event code: T3

    PLBE No opposition filed within time limit

    Free format text: ORIGINAL CODE: 0009261

    STAA Information on the status of an ep patent application or granted ep patent

    Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

    26N No opposition filed
    REG Reference to a national code

    Ref country code: GB

    Ref legal event code: IF02

    PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

    Ref country code: DK

    Payment date: 20090824

    Year of fee payment: 20

    PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

    Ref country code: CH

    Payment date: 20090824

    Year of fee payment: 20

    Ref country code: SE

    Payment date: 20090824

    Year of fee payment: 20

    Ref country code: LU

    Payment date: 20090824

    Year of fee payment: 20

    Ref country code: GB

    Payment date: 20090820

    Year of fee payment: 20

    PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

    Ref country code: ES

    Payment date: 20091007

    Year of fee payment: 20

    Ref country code: DE

    Payment date: 20090826

    Year of fee payment: 20

    Ref country code: AT

    Payment date: 20090824

    Year of fee payment: 20

    PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

    Ref country code: BE

    Payment date: 20090827

    Year of fee payment: 20

    Ref country code: NL

    Payment date: 20090827

    Year of fee payment: 20

    PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

    Ref country code: IT

    Payment date: 20090827

    Year of fee payment: 20

    REG Reference to a national code

    Ref country code: NL

    Ref legal event code: V4

    Effective date: 20101004

    REG Reference to a national code

    Ref country code: CH

    Ref legal event code: PL

    REG Reference to a national code

    Ref country code: DK

    Ref legal event code: EUP

    REG Reference to a national code

    Ref country code: GB

    Ref legal event code: PE20

    Expiry date: 20101003

    BE20 Be: patent expired

    Owner name: *OPTEIN INC.

    Effective date: 20101004

    EUG Se: european patent has lapsed
    PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

    Ref country code: NL

    Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

    Effective date: 20101004

    PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

    Ref country code: GB

    Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

    Effective date: 20101003

    PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

    Ref country code: FR

    Payment date: 20090907

    Year of fee payment: 20

    REG Reference to a national code

    Ref country code: ES

    Ref legal event code: FD2A

    Effective date: 20120511

    PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

    Ref country code: ES

    Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

    Effective date: 20101005

    PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

    Ref country code: DE

    Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

    Effective date: 20101004